1
|
Tsuji RK, Hamerschmidt R, Lavinsky J, Felix F, Silva VAR. Brazilian Society of Otology task force - cochlear implant ‒ recommendations based on strength of evidence. Braz J Otorhinolaryngol 2025; 91:101512. [PMID: 39442262 PMCID: PMC11539123 DOI: 10.1016/j.bjorl.2024.101512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 09/02/2024] [Indexed: 10/25/2024] Open
Abstract
OBJECTIVE To make evidence-based recommendations for the indications and complications of Cochlear Implant (CI) surgery in adults and children. METHODS Task force members were educated on knowledge synthesis methods, including electronic database search, review and selection of relevant citations, and critical appraisal of selected studies. Articles written in English or Portuguese on cochlear implantation were eligible for inclusion. The American College of Physicians' guideline grading system and the American Thyroid Association's guideline criteria were used for critical appraisal of evidence and recommendations for therapeutic interventions. RESULTS The topics were divided into 2 parts: (1) Evaluation of candidate patients and indications for CI surgery; (2) CI surgery - techniques and complications. CONCLUSIONS CI is a safe device for auditory rehabilitation of patients with severe-to-profound hearing loss. In recent years, indications for unilateral hearing loss and vestibular schwannoma have been expanded, with encouraging results. However, for a successful surgery, commitment of family members and patients in the hearing rehabilitation process is essential.
Collapse
Affiliation(s)
- Robinson Koji Tsuji
- Universidade de São Paulo (USP), Faculdade de Medicina, Departamento de Otorrinolaringologia, São Paulo, SP, Brazil
| | - Rogério Hamerschmidt
- Universidade Federal do Paraná (UFPR), Departamento de Otorrinolaringologia, Curitiba, PR, Brazil
| | - Joel Lavinsky
- Universidade Federal do Rio Grande do Sul (UFRGS), Departamento de Ciências Morfológicas, Porto Alegre, RS, Brazil
| | - Felippe Felix
- Universidade Federal do Rio de Janeiro (UFRJ), Hospital Universitário Clementino Fraga Filho (HUCFF), Rio de Janeiro, RJ, Brazil
| | - Vagner Antonio Rodrigues Silva
- Universidade de Campinas (Unicamp), Faculdade de Ciências Médicas (FCM), Departamento de Otorrinolaringologia e Cirurgia de Cabeça e Pescoço, Campinas, SP, Brazil.
| |
Collapse
|
2
|
Carlyon RP, Deeks JM, Delgutte B, Chung Y, Vollmer M, Ohl FW, Kral A, Tillein J, Litovsky RY, Schnupp J, Rosskothen-Kuhl N, Goldsworthy RL. Limitations on Temporal Processing by Cochlear Implant Users: A Compilation of Viewpoints. Trends Hear 2025; 29:23312165251317006. [PMID: 40095543 DOI: 10.1177/23312165251317006] [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] [Indexed: 03/19/2025] Open
Abstract
Cochlear implant (CI) users are usually poor at using timing information to detect changes in either pitch or sound location. This deficit occurs even for listeners with good speech perception and even when the speech processor is bypassed to present simple, idealized stimuli to one or more electrodes. The present article presents seven expert opinion pieces on the likely neural bases for these limitations, the extent to which they are modifiable by sensory experience and training, and the most promising ways to overcome them in future. The article combines insights from physiology and psychophysics in cochlear-implanted humans and animals, highlights areas of agreement and controversy, and proposes new experiments that could resolve areas of disagreement.
Collapse
Affiliation(s)
- Robert P Carlyon
- Cambridge Hearing Group, MRC Cognition & Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - John M Deeks
- Cambridge Hearing Group, MRC Cognition & Brain Sciences Unit, University of Cambridge, Cambridge, UK
| | - Bertrand Delgutte
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA, USA
| | - Yoojin Chung
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA, USA
| | - Maike Vollmer
- Department of Experimental Audiology, University Clinic of Otolaryngology, Head and Neck Surgery, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Frank W Ohl
- Leibniz Institute for Neurobiology (LIN), Magdeburg, Germany
| | - Andrej Kral
- Institute of Audio-Neuro-Technology & Department of Experimental Otology, Clinics of Otolaryngology, Head and Neck Surgery, Hannover Medical School, Hannover, Germany
| | - Jochen Tillein
- Clinics of Otolaryngology, Head and Neck Surgery, J.W.Goethe University, Frankfurt, Germany
- MedEl Company, Hannover, Germany
| | - Ruth Y Litovsky
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Jan Schnupp
- Gerald Choa Neuroscience Institute and Department of Otolaryngology, Chinese University of Hong Kong, Hong Kong (NB Hong Kong is a Special Administrative Region) of China
| | - Nicole Rosskothen-Kuhl
- Neurobiological Research Laboratory, Section for Experimental and Clinical Otology, Department of Oto-Rhino-Laryngology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Bernstein Center Freiburg & Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Raymond L Goldsworthy
- Auditory Research Center, Caruso Department of Otolaryngology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
3
|
Wu M, Wang Y, Zhao X, Xin T, Wu K, Liu H, Wu S, Liu M, Chai X, Li J, Wei C, Zhu C, Liu Y, Zhang YX. Anti-phasic oscillatory development for speech and noise processing in cochlear implanted toddlers. Child Dev 2024; 95:1693-1708. [PMID: 38742715 DOI: 10.1111/cdev.14105] [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] [Indexed: 05/16/2024]
Abstract
Human brain demonstrates amazing readiness for speech and language learning at birth, but the auditory development preceding such readiness remains unknown. Cochlear implanted (CI) children (n = 67; mean age 2.77 year ± 1.31 SD; 28 females) with prelingual deafness provide a unique opportunity to study this stage. Using functional near-infrared spectroscopy, it was revealed that the brain of CI children was irresponsive to sounds at CI hearing onset. With increasing CI experiences up to 32 months, the brain demonstrated function, region and hemisphere specific development. Most strikingly, the left anterior temporal lobe showed an oscillatory trajectory, changing in opposite phases for speech and noise. The study provides the first longitudinal brain imaging evidence for early auditory development preceding speech acquisition.
Collapse
Affiliation(s)
- Meiyun Wu
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Yuyang Wang
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Department of Otolaryngology Head and Neck Surgery, Hunan Provincial People's Hospital (First Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Xue Zhao
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Tianyu Xin
- Department of Otolaryngology Head and Neck Surgery, Peking University First Hospital, Beijing, China
| | - Kun Wu
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Haotian Liu
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Department of Otolaryngology Head and Neck Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Shinan Wu
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Min Liu
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Xiaoke Chai
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Jinhong Li
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Chaogang Wei
- Department of Otolaryngology Head and Neck Surgery, Peking University First Hospital, Beijing, China
| | - Chaozhe Zhu
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Yuhe Liu
- Department of Otolaryngology Head and Neck Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yu-Xuan Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning & IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| |
Collapse
|
4
|
Gansemer BM, Rahman MT, Zhang Z, Green SH. Spiral ganglion neuron degeneration in aminoglycoside-deafened rats involves innate and adaptive immune responses not requiring complement. Front Mol Neurosci 2024; 17:1389816. [PMID: 38840777 PMCID: PMC11151750 DOI: 10.3389/fnmol.2024.1389816] [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: 02/22/2024] [Accepted: 04/29/2024] [Indexed: 06/07/2024] Open
Abstract
Spiral ganglion neurons (SGNs) transmit auditory information from cochlear hair cells to the brain. SGNs are thus not only important for normal hearing, but also for effective functioning of cochlear implants, which stimulate SGNs when hair cells are missing. SGNs slowly degenerate following aminoglycoside-induced hair cell loss, a process thought to involve an immune response. However, the specific immune response pathways involved remain unknown. We used RNAseq to gain a deeper understanding immune-related and other transcriptomic changes that occur in the rat spiral ganglion after kanamycin-induced deafening. Among the immune and inflammatory genes that were selectively upregulated in deafened spiral ganglia, the complement cascade genes were prominent. We then assessed SGN survival, as well as immune cell numbers and activation, in the spiral ganglia of rats with a CRISPR-Cas9-mediated knockout of complement component 3 (C3). Similar to previous findings in our lab and other deafened rodent models, we observed an increase in macrophage number and increased expression of CD68, a marker of phagocytic activity and cell activation, in macrophages in the deafened ganglia. Moreover, we found an increase in MHCII expression on spiral ganglion macrophages and an increase in lymphocyte number in the deafened ganglia, suggestive of an adaptive immune response. However, C3 knockout did not affect SGN survival or increase in macrophage number/activation, implying that complement activation does not play a role in SGN death after deafening. Together, these data suggest that both innate and adaptive immune responses are activated in the deafened spiral ganglion, with the adaptive response directly contributing to cochlear neurodegeneration.
Collapse
Affiliation(s)
| | | | | | - Steven H. Green
- Department of Biology, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
5
|
Seicol BJ, Guo Z, Garrity K, Xie R. Potential uses of auditory nerve stimulation to modulate immune responses in the inner ear and auditory brainstem. Front Integr Neurosci 2023; 17:1294525. [PMID: 38162822 PMCID: PMC10755874 DOI: 10.3389/fnint.2023.1294525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024] Open
Abstract
Bioelectronic medicine uses electrical stimulation of the nervous system to improve health outcomes throughout the body primarily by regulating immune responses. This concept, however, has yet to be applied systematically to the auditory system. There is growing interest in how cochlear damage and associated neuroinflammation may contribute to hearing loss. In conjunction with recent findings, we propose here a new perspective, which could be applied alongside advancing technologies, to use auditory nerve (AN) stimulation to modulate immune responses in hearing health disorders and following surgeries for auditory implants. In this article we will: (1) review the mechanisms of inflammation in the auditory system in relation to various forms of hearing loss, (2) explore nerve stimulation to reduce inflammation throughout the body and how similar neural-immune circuits likely exist in the auditory system (3) summarize current methods for stimulating the auditory system, particularly the AN, and (4) propose future directions to use bioelectronic medicine to ameliorate harmful immune responses in the inner ear and auditory brainstem to treat refractory conditions. We will illustrate how current knowledge from bioelectronic medicine can be applied to AN stimulation to resolve inflammation associated with implantation and disease. Further, we suggest the necessary steps to get discoveries in this emerging field from bench to bedside. Our vision is a future for AN stimulation that includes additional protocols as well as advances in devices to target and engage neural-immune circuitry for therapeutic benefits.
Collapse
Affiliation(s)
- Benjamin J. Seicol
- Department of Otolaryngology, The Ohio State University, Columbus, OH, United States
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| | - Zixu Guo
- Department of Otolaryngology, The Ohio State University, Columbus, OH, United States
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| | - Katy Garrity
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| | - Ruili Xie
- Department of Otolaryngology, The Ohio State University, Columbus, OH, United States
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
6
|
Garcia A, Haleem A, Chari DA, Morse-Fortier C, Arenberg JG, Lee DJ. Influence of listening environment on usage patterns in cochlear implant patients with single-sided deafness. Cochlear Implants Int 2023; 24:335-341. [PMID: 36846887 DOI: 10.1080/14670100.2023.2176990] [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] [Indexed: 03/01/2023]
Abstract
OBJECTIVE To compare cochlear implant (CI) data logging of patients with single-sided deafness (SSD) and bilateral sensorineural hearing loss (biSNHL) in various acoustic environments and study the implications of data logging on auditory performance. STUDY DESIGN Retrospective case control study. METHODS Adult CI patients with SSD or biSNHL from 2010 to 2021 with usage data collected at 3-, 6-, and 12-months following device activation were identified. The CI listening environment was defined as speech in noise, speech in quiet, quiet, music or noise. Auditory performance was measured using the CNC word, AzBio sentence tests and the Tinnitus Handicap Index (THI). RESULTS 60 adults with SSD or biSNHL were included. CI patients with biSNHL wore their devices more than those with SSD at 3-months post-activation (11.18 versus 8.97 hours/day, p = 0.04), though there were no significant differences at 6-12 months. Device usage was highest in the speech in quiet environment. In SSD CI users, there was a positive correlation (p = 0.03) between device use and CNC scores at 12-months and an improvement in THI scores at 12-months (p = 0.0004). CONCLUSIONS CI users with SSD and biSNHL have comparable duration of device usage at longer follow-up periods with greatest device usage recorded in speech in quiet environments.
Collapse
Affiliation(s)
- Alejandro Garcia
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Harvard Medical School, 243 Charles Street, Boston, MA, USA
- Eaton Peabody Laboratories (EPL), Massachusetts Eye and Ear, Harvard Medical School, 243 Charles Street, Boston, MA, USA
| | - Afash Haleem
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Harvard Medical School, 243 Charles Street, Boston, MA, USA
- Eaton Peabody Laboratories (EPL), Massachusetts Eye and Ear, Harvard Medical School, 243 Charles Street, Boston, MA, USA
| | - Divya A Chari
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Harvard Medical School, 243 Charles Street, Boston, MA, USA
- Eaton Peabody Laboratories (EPL), Massachusetts Eye and Ear, Harvard Medical School, 243 Charles Street, Boston, MA, USA
| | - Charlotte Morse-Fortier
- Department of Audiology, Massachusetts Eye and Ear, 243 Charles Street, Boston, MA, USA
- Eaton Peabody Laboratories (EPL), Massachusetts Eye and Ear, Harvard Medical School, 243 Charles Street, Boston, MA, USA
| | - Julie G Arenberg
- Department of Audiology, Massachusetts Eye and Ear, 243 Charles Street, Boston, MA, USA
- Eaton Peabody Laboratories (EPL), Massachusetts Eye and Ear, Harvard Medical School, 243 Charles Street, Boston, MA, USA
| | - Daniel J Lee
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Harvard Medical School, 243 Charles Street, Boston, MA, USA
- Eaton Peabody Laboratories (EPL), Massachusetts Eye and Ear, Harvard Medical School, 243 Charles Street, Boston, MA, USA
| |
Collapse
|
7
|
Berninger E, Drott M, Romanitan M, Tranebjærg L, Hellström S. Congenital Nonprofound Bilateral Sensorineural Hearing Loss in Children: Comprehensive Characterization of Auditory Function and Hearing Aid Benefit. Audiol Res 2022; 12:539-563. [PMID: 36285911 PMCID: PMC9598400 DOI: 10.3390/audiolres12050054] [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: 08/26/2022] [Revised: 09/28/2022] [Accepted: 10/02/2022] [Indexed: 11/25/2022] Open
Abstract
A prospective cross-sectional design was used to characterize congenital bilateral sensorineural hearing loss (SNHL). The underlying material of >30,000 consecutively screened newborns comprised 11 subjects with nonprofound, alleged nonsyndromic, SNHL. Comprehensive audiological testing was performed at ≈11 years of age. Results showed symmetrical sigmoid-like median pure-tone thresholds (PTTs) reaching 50−60 dB HL. The congenital SNHL revealed recruitment, increased upward spread of masking, distortion product otoacoustic emission (DPOAE) dependent on PTT (≤60 dB HL), reduced auditory brainstem response (ABR) amplitude, and normal magnetic resonance imaging. Unaided recognition of speech in spatially separate competing speech (SCS) deteriorated with increasing uncomfortable loudness level (UCL), plausibly linked to reduced afferent signals. Most subjects demonstrated hearing aid (HA) benefit in a demanding laboratory listening situation. Questionnaires revealed HA benefit in real-world listening situations. This functional characterization should be important for the outline of clinical guidelines. The distinct relationship between DPOAE and PTT, up to the theoretical limit of cochlear amplification, and the low ABR amplitude remain to be elucidated. The significant relation between UCL and SCS has implications for HA-fitting. The fitting of HAs based on causes, mechanisms, and functional characterization of the SNHL may be an individualized intervention approach and deserves future research.
Collapse
Affiliation(s)
- Erik Berninger
- Department of Clinical Science, Intervention and Technology, Division of Ear, Nose and Throat Diseases, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Audiology and Neurotology, Karolinska University Hospital, 141 86 Stockholm, Sweden
- Correspondence: or
| | - Maria Drott
- Department of Clinical Science, Intervention and Technology, Division of Ear, Nose and Throat Diseases, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Audiology and Neurotology, Karolinska University Hospital, 141 86 Stockholm, Sweden
| | - Mircea Romanitan
- Department of Clinical Science, Intervention and Technology, Division of Ear, Nose and Throat Diseases, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Audiology and Neurotology, Karolinska University Hospital, 141 86 Stockholm, Sweden
| | - Lisbeth Tranebjærg
- Department of Clinical Genetics, The University Hospital Rigshospital/The Kennedy Centre, DK-2600 Copenhagen, Denmark
- Institute of Clinical Medicine, University of Copenhagen, DK-1165 Copenhagen, Denmark
| | - Sten Hellström
- Department of Clinical Science, Intervention and Technology, Division of Ear, Nose and Throat Diseases, Karolinska Institutet, 171 77 Stockholm, Sweden
- Department of Audiology and Neurotology, Karolinska University Hospital, 141 86 Stockholm, Sweden
| |
Collapse
|
8
|
Seicol BJ, Lin S, Xie R. Age-Related Hearing Loss Is Accompanied by Chronic Inflammation in the Cochlea and the Cochlear Nucleus. Front Aging Neurosci 2022; 14:846804. [PMID: 35418849 PMCID: PMC8995794 DOI: 10.3389/fnagi.2022.846804] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/07/2022] [Indexed: 12/24/2022] Open
Abstract
Age-related hearing loss (ARHL) is a major hearing impairment characterized by pathological changes in both the peripheral and central auditory systems. Low-grade inflammation was observed in the cochlea of deceased human subjects with ARHL and animal models of early onset ARHL, which suggests that inflammation contributes to the development of ARHL. However, it remains elusive how chronic inflammation progresses during normal aging in the cochlea, and especially the accompanying changes of neuroinflammation in the central auditory system. To address this, we investigated chronic inflammation in both the cochlea and the cochlear nucleus (CN) of CBA/CaJ mice, an inbred mouse strain that undergoes normal aging and develops human, like-late-onset ARHL. Using immunohistochemistry, confocal microscopy, and quantitative image processing, we measured the accumulation and activation of macrophages in the cochlea and microglia in the CN using their shared markers: ionized calcium binding adaptor molecule 1 (Iba1) and CD68-a marker of phagocytic activity. We found progressive increases in the area covered by Iba1-labeled macrophages and enhanced CD68 staining in the osseous spiral lamina of the cochlea that correlated with elevated ABR threshold across the lifespan. During the process, we further identified significant increases in microglial activation and C1q deposition in the CN, indicating increased neuroinflammation and complement activation in the central auditory system. Our study suggests that during normal aging, chronic inflammation occurs in both the peripheral and the central auditory system, which may contribute in coordination to the development of ARHL.
Collapse
Affiliation(s)
- Benjamin J. Seicol
- Department of Otolaryngology—Head and Neck Surgery, The Ohio State University, Columbus, OH, United States
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| | - Shengyin Lin
- Department of Otolaryngology—Head and Neck Surgery, The Ohio State University, Columbus, OH, United States
| | - Ruili Xie
- Department of Otolaryngology—Head and Neck Surgery, The Ohio State University, Columbus, OH, United States
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
9
|
Impact of Duration of Deafness on Speech Perception in Single-Sided Deafness Cochlear Implantation in Adults. Otol Neurotol 2022; 43:e45-e49. [PMID: 34889841 DOI: 10.1097/mao.0000000000003357] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To evaluate the impact of prolonged auditory deprivation on speech perception outcomes in adult acquired single-sided deafness (SSD) cochlear implant (CI) recipients. STUDY DESIGN Retrospective case series. SETTING Tertiary care academic center. PATIENTS Acquired SSD in adults with and without prolonged duration of deafness (defined as >10 yr) who underwent CI between 2014 and 2019. INTERVENTIONS CI. MAIN OUTCOME MEASURES Consonant-nucleus-consonant (CNC) and AzBio in quiet scores within first year of follow-up. RESULTS A total of 35 adult patients with SSD were evaluated, with a median overall duration of deafness of 2.4 years (interquartile range [IQR] 1.2-6.0 yr): seven patients with prolonged auditory deprivation (median 18 yr, IQR 15-28) were compared with 28 SSD patients with duration of deafness less than 10 years (median 1.7 yr, IQR 1.1-3.2). At last follow-up, the median CNC scores were 39% (IQR 31-64) and 54% (IQR 46-64) for the prolonged and shorter duration of deafness cohorts, respectively (p = 0.3). The median AzBio scores were 66% (IQR 65-68) and 72% (IQR 60-82) for the prolonged and shorter duration of deafness cohorts, respectively (p = 0.6). In a separate analysis evaluating duration of deafness as a continuous variable across all 35 patients, Spearman correlation coefficients for associations of duration of deafness with most recent CNC and AzBio scores were -0.02 (p = 0.9) and 0.02 (p = 0.9), respectively. CONCLUSIONS Adult CI recipients with acquired SSD, with and without prolonged auditory deprivation, demonstrated comparable speech perception scores. Prolonged duration of deafness alone should not preclude a motivated SSD patient from undergoing cochlear implantation.
Collapse
|
10
|
Establishing Reproducibility and Correlation of Cochlear Microphonic Amplitude to Implant Electrode Position Using Intraoperative Electrocochleography and Postoperative Cone Beam Computed Tomography. Ear Hear 2021; 42:1263-1275. [PMID: 33813521 PMCID: PMC8378545 DOI: 10.1097/aud.0000000000001010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Supplemental Digital Content is available in the text. Objectives: The primary objective of this study was to establish the reproducibility of cochlear microphonic (CM) recordings obtained from a cochlear implant (CI) electrode contact during and immediately after insertion. This was achieved by evaluating the insertion angle and calculating the position of the apical electrode contact during insertion, using postoperative cone beam computed tomography (CBCT). The secondary objective was to create individualized patient maps of electrode contacts located within acoustically sensitive regions by correlating the CM amplitude to the electrode position determined using CBCT. Methods: CMs were recorded from a CI electrode contact during and immediately after insertion in 12 patients (n = 14 ears). Intraoperative recordings were made for a 0.5 kHz tone burst stimulus and were recorded from the apical electrode contact. Postinsertion recordings were made from the odd-numbered electrode contacts (1–15) along the array, using a range of stimulus frequencies (from 0.125 to 2 kHz). The time point at which each electrode contact passed through the round window was noted throughout the insertion, and the CM amplitude at this point was correlated to postoperative CBCT. This correlation was then used to estimate the CM amplitude at particular points within the cochlea, which was in turn compared with the amplitudes recorded from each electrode postoperatively to assess the reproducibility of the recordings. Results: Significant correlation was shown between intraoperative insertion and postinsertion angles at two amplitude events (maximum amplitude: 29° mean absolute error, r = 0.77, p = 0.006; 10% of maximum amplitude: 52° mean absolute error, r = 0.85, p = 0.002). Conclusion: We have developed a novel method to demonstrate the reproducibility of the CM responses recorded from a CI electrode during insertion. By correlating the CM amplitude with the postoperative CBCT, we have also been able to create individualized maps of CM responses, categorizing the cochlea into acoustically responsive and unresponsive regions. If the electrode contacts within the acoustically sensitive regions are shown to be associated with improved loudness discrimination, it could have implications for optimal electrode mapping and placement.
Collapse
|
11
|
Hintze A, Gültas M, Semmelhack EA, Wichmann C. Ultrastructural maturation of the endbulb of Held active zones comparing wild-type and otoferlin-deficient mice. iScience 2021; 24:102282. [PMID: 33851098 PMCID: PMC8022229 DOI: 10.1016/j.isci.2021.102282] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/18/2021] [Accepted: 03/03/2021] [Indexed: 11/13/2022] Open
Abstract
Endbulbs of Held are located in the anteroventral cochlear nucleus and present the first central synapses of the auditory pathway. During development, endbulbs mature functionally to enable rapid and powerful synaptic transmission with high temporal precision. This process is accompanied by morphological changes of endbulb terminals. Loss of the hair cell-specific protein otoferlin (Otof) abolishes neurotransmission in the cochlea and results in the smaller endbulb of Held terminals. Thus, peripheral hearing impairment likely also leads to alterations in the morphological synaptic vesicle (SV) pool size at individual endbulb of Held active zones (AZs). Here, we investigated endbulb AZs in pre-hearing, young, and adult wild-type and Otof−/− mice. During maturation, SV numbers at endbulb AZs increased in wild-type mice but were found to be reduced in Otof−/− mice. The SV population at a distance of 0–15 nm was most strongly affected. Finally, overall SV diameters decreased in Otof−/− animals during maturation. Maturation of wt endbulb of Held active zones leads to more synaptic vesicles At endbulbs of otoferlin knockout mice, synaptic vesicles decline with age Mainly two distinct synaptic vesicle populations are affected Synaptic vesicles sizes are reduced in six-month-old otoferlin knockout animals
Collapse
Affiliation(s)
- Anika Hintze
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience, InnerEarLab and Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany.,Collaborative Research Center 1286, University of Göttingen, Göttingen, Germany.,Göttingen Graduate School for Neurosciences, Biophysics and Molecular Biosciences, University of Göttingen, Göttingen, Germany
| | - Mehmet Gültas
- Breeding Informatics Group, Department of Animal Sciences, Georg-August-University Göttingen, Göttingen, Germany
| | - Esther A Semmelhack
- Developmental, Neural, and Behavioral Biology MSc/PhD Program, University of Göttingen, Göttingen, Germany
| | - Carolin Wichmann
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience, InnerEarLab and Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany.,Collaborative Research Center 1286, University of Göttingen, Göttingen, Germany
| |
Collapse
|
12
|
Purcell PL, Deep NL, Waltzman SB, Roland JT, Cushing SL, Papsin BC, Gordon KA. Cochlear Implantation in Infants: Why and How. Trends Hear 2021; 25:23312165211031751. [PMID: 34281434 PMCID: PMC8295935 DOI: 10.1177/23312165211031751] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 04/22/2021] [Accepted: 06/23/2021] [Indexed: 11/25/2022] Open
Abstract
In children with congenital deafness, cochlear implantation (CI) prior to 12 months of age offers the opportunity to foster more typical auditory development during late infancy and early childhood. Recent studies have found a positive association between early implantation and expressive and receptive language outcomes, with some children able to achieve normal language skills by the time of school entry. Universal newborn hearing screening improved early detection and diagnosis of congenital hearing loss, allowing for earlier intervention, including decision-making regarding cochlear implant (CI) candidacy. It can be more challenging to confirm CI candidacy in infants; therefore, a multidisciplinary approach, including objective audiometric testing, is recommended to not only confirm the diagnosis but also to counsel families regarding expectations and long-term management. Surgeons performing CI surgery in young children should consider both the anesthetic risks of surgery in infancy and the ways in which mastoid anatomy may differ between infants and older children or adults. Multiple studies have found CI surgery in infants can be performed safely and effectively. This article reviews current evidence regarding indications for implantation in children younger than 12 months of age and discusses perioperative considerations and surgical technique.
Collapse
Affiliation(s)
- Patricia L. Purcell
- Department of Otolaryngology, Head & Neck Surgery, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Nicholas L. Deep
- Department of Otolaryngology, Head & Neck Surgery, New York University Grossman School of Medicine, New York, New York, United States
| | - Susan B. Waltzman
- Department of Otolaryngology, Head & Neck Surgery, New York University Grossman School of Medicine, New York, New York, United States
| | - J. Thomas Roland
- Department of Otolaryngology, Head & Neck Surgery, New York University Grossman School of Medicine, New York, New York, United States
| | - Sharon L. Cushing
- Department of Otolaryngology, Head & Neck Surgery, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Blake C. Papsin
- Department of Otolaryngology, Head & Neck Surgery, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Karen A. Gordon
- Department of Otolaryngology, Head & Neck Surgery, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
13
|
Eklöf M, Asp F, Berninger E. Sound localization latency in normal hearing and simulated unilateral hearing loss. Hear Res 2020; 395:108011. [PMID: 32792116 DOI: 10.1016/j.heares.2020.108011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 05/14/2020] [Accepted: 05/28/2020] [Indexed: 10/24/2022]
Abstract
Directing gaze towards auditory events is a natural behavior. In addition to the well-known accuracy of auditory elicited gaze responses for normal binaural listening, their latency is a measure of possible clinical interest and methodological importance. The aim was to develop a clinically feasible method to assess sound localization latency (SLL), and to study SLL as a function of simulated unilateral hearing loss (SUHL) and the relationship with accuracy. Eight healthy and normal-hearing adults (18-40 years) participated in this study. Horizontal gaze responses, recorded by non-invasive corneal reflection eye-tracking, were obtained during azimuthal shifts (24 trials) of a 3-min continuous auditory stimulus. In each trial, a sigmoid function was fitted to gaze samples. Latency was estimated by the abscissa corresponding to 50% of the arctangent amplitude. SLL was defined as the mean latency across trials. SLL was measured in normal-hearing and simulated SUHL conditions (SUHL30 and SUHL43: mean threshold of 30 dB HL and 43 dB HL across 0.5, 1, 2, and 4 kHz). In the normal-hearing condition, the mean ± SD SLL was 280 ± 40 ms (n = 8) with a test-retest SD = 20 ms. A linear mixed model showed a statistically significant effect of listening condition on SLL. The SUHL30 and SUHL43 conditions revealed a mean SLL of 370 ± 49 ms and 540 ± 120 ms, respectively. Repeated measures correlation analysis showed a clear relationship between SLL and the average sound localization accuracy (R2 = 0.94). The rapid and reliable method to obtain SLL may be an important clinical tool for evaluation of binaural processing. Future studies in clinical cohorts are needed to assess whether SLL may reveal information about binaural processing abilities beyond that afforded by sound localization accuracy.
Collapse
Affiliation(s)
- Martin Eklöf
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden; Department of ENT, Section of Hearing Implants, Karolinska University Hospital, Stockholm, Sweden.
| | - Filip Asp
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden; Department of ENT, Section of Hearing Implants, Karolinska University Hospital, Stockholm, Sweden
| | - Erik Berninger
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden; Department of Audiology and Neurotology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
14
|
Scheffel JL, Mohammed SS, Borcean CK, Parng AJ, Yoon HJ, Gutierrez DA, Yu WM. Spatiotemporal Analysis of Cochlear Nucleus Innervation by Spiral Ganglion Neurons that Serve Distinct Regions of the Cochlea. Neuroscience 2020; 446:43-58. [PMID: 32866604 DOI: 10.1016/j.neuroscience.2020.08.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/21/2020] [Accepted: 08/21/2020] [Indexed: 10/23/2022]
Abstract
Cochlear neurons innervate the brainstem cochlear nucleus in a tonotopic fashion according to their sensitivity to different sound frequencies (known as the neuron's characteristic frequency). It is unclear whether these neurons with distinct characteristic frequencies use different strategies to innervate the cochlear nucleus. Here, we use genetic approaches to differentially label spiral ganglion neurons (SGNs) and their auditory nerve fibers (ANFs) that relay different characteristic frequencies in mice. We found that SGN populations that supply distinct regions of the cochlea employ different cellular strategies to target and innervate neurons in the cochlear nucleus during tonotopic map formation. ANFs that will exhibit high-characteristic frequencies initially overshoot and sample a large area of targets before refining their connections to correct targets, while fibers that will exhibit low-characteristic frequencies are more accurate in initial targeting and undergo minimal target sampling. Moreover, similar to their peripheral projections, the central projections of ANFs show a gradient of development along the tonotopic axis, with outgrowth and branching of prospective high-frequency ANFs initiated about two days earlier than those of prospective low-frequency ANFs. The processes of synaptogenesis are similar between high- and low-frequency ANFs, but a higher proportion of low-frequency ANFs form smaller endbulb synaptic endings. These observations reveal the diversity of cellular mechanisms that auditory neurons that will become functionally distinct use to innervate their targets during tonotopic map formation.
Collapse
Affiliation(s)
- Jennifer L Scheffel
- Department of Biology, Loyola University Chicago, Chicago, IL 60660, United States
| | - Samiha S Mohammed
- Department of Biology, Loyola University Chicago, Chicago, IL 60660, United States
| | - Chloe K Borcean
- Department of Biology, Loyola University Chicago, Chicago, IL 60660, United States
| | - Annie J Parng
- Department of Biology, Loyola University Chicago, Chicago, IL 60660, United States
| | - Hyun Ju Yoon
- Department of Biology, Loyola University Chicago, Chicago, IL 60660, United States
| | - Darwin A Gutierrez
- Department of Biology, Loyola University Chicago, Chicago, IL 60660, United States
| | - Wei-Ming Yu
- Department of Biology, Loyola University Chicago, Chicago, IL 60660, United States.
| |
Collapse
|
15
|
Data logging variables and speech perception in prelingually deafened pediatric cochlear implant users. Int J Pediatr Otorhinolaryngol 2020; 133:110003. [PMID: 32203760 DOI: 10.1016/j.ijporl.2020.110003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 11/23/2022]
Abstract
OBJECTIVES To investigate the relationship among objectively gathered data logging measurements, patient-related variables, and speech recognition performance of pediatric CI users. METHODS AND MATERIALS Thirty-two prelingually implanted children who have the ability to perform word discrimination test were included in this study. To reveal the relationship between speech perception abilities and auditory exposure, seven data logging variables were analyzed: "on-air," "off-air," "coil-off," "speech," "speech in noise," "music" and "noise. In addition, implantation age (months) and CI usage duration (months) were taken into account. Finally, it was investigated the differences between unilateral, sequential bilateral, and simultaneous bilateral CI users in terms of all study variables. RESULTS The average on-air time ranged between 10.52 and 12.30 in the groups. In the case of sequential implantation, smaller on-air and higher coil off values were observed with the second CI. In the case of simultaneous bilateral implantation, data logging measurements were almost the same in both implants. WRS was significantly correlated (p < 0.05) with on-air time (r = 0.62), coil-off count (r = -0.48), chronological age (r = 0.48), and CI duration (r = 0.44). Multiple linear regression model was fit to predict the WRS, with on-air time, CI duration, and chronological age as predictors. CONCLUSIONS The critical importance of early intervention and long-term use of CI is well-established in the literature and is also corroborated by our findings. However, the key findings of the present study are that consistent CI use and the quality of daily listening environment also exerted a major and positive effect on the speech recognition performance of pediatric CI users. Therefore, during the monitoring of pediatric CI recipients, it is important to know the device usage data in order to detect problems in the early stages after CI.
Collapse
|
16
|
Persic D, Thomas ME, Pelekanos V, Ryugo DK, Takesian AE, Krumbholz K, Pyott SJ. Regulation of auditory plasticity during critical periods and following hearing loss. Hear Res 2020; 397:107976. [PMID: 32591097 PMCID: PMC8546402 DOI: 10.1016/j.heares.2020.107976] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/15/2020] [Accepted: 04/14/2020] [Indexed: 02/07/2023]
Abstract
Sensory input has profound effects on neuronal organization and sensory maps in the brain. The mechanisms regulating plasticity of the auditory pathway have been revealed by examining the consequences of altered auditory input during both developmental critical periods—when plasticity facilitates the optimization of neural circuits in concert with the external environment—and in adulthood—when hearing loss is linked to the generation of tinnitus. In this review, we summarize research identifying the molecular, cellular, and circuit-level mechanisms regulating neuronal organization and tonotopic map plasticity during developmental critical periods and in adulthood. These mechanisms are shared in both the juvenile and adult brain and along the length of the auditory pathway, where they serve to regulate disinhibitory networks, synaptic structure and function, as well as structural barriers to plasticity. Regulation of plasticity also involves both neuromodulatory circuits, which link plasticity with learning and attention, as well as ascending and descending auditory circuits, which link the auditory cortex and lower structures. Further work identifying the interplay of molecular and cellular mechanisms associating hearing loss-induced plasticity with tinnitus will continue to advance our understanding of this disorder and lead to new approaches to its treatment. During CPs, brain plasticity is enhanced and sensitive to acoustic experience. Enhanced plasticity can be reinstated in the adult brain following hearing loss. Molecular, cellular, and circuit-level mechanisms regulate CP and adult plasticity. Plasticity resulting from hearing loss may contribute to the emergence of tinnitus. Modifying plasticity in the adult brain may offer new treatments for tinnitus.
Collapse
Affiliation(s)
- Dora Persic
- University of Groningen, University Medical Center Groningen, Groningen, Department of Otorhinolaryngology and Head/Neck Surgery, 9713, GZ, Groningen, the Netherlands
| | - Maryse E Thomas
- Eaton-Peabody Laboratories, Massachusetts Eye & Ear and Department of Otorhinolaryngology and Head/Neck Surgery, Harvard Medical School, Boston, MA, USA
| | - Vassilis Pelekanos
- Hearing Sciences, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, University Park, Nottingham, UK
| | - David K Ryugo
- Hearing Research, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia; School of Medical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia; Department of Otolaryngology, Head, Neck & Skull Base Surgery, St Vincent's Hospital, Sydney, NSW, 2010, Australia
| | - Anne E Takesian
- Eaton-Peabody Laboratories, Massachusetts Eye & Ear and Department of Otorhinolaryngology and Head/Neck Surgery, Harvard Medical School, Boston, MA, USA
| | - Katrin Krumbholz
- Hearing Sciences, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, University Park, Nottingham, UK
| | - Sonja J Pyott
- University of Groningen, University Medical Center Groningen, Groningen, Department of Otorhinolaryngology and Head/Neck Surgery, 9713, GZ, Groningen, the Netherlands.
| |
Collapse
|
17
|
Ratnanather JT. Structural neuroimaging of the altered brain stemming from pediatric and adolescent hearing loss-Scientific and clinical challenges. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2020; 12:e1469. [PMID: 31802640 PMCID: PMC7307271 DOI: 10.1002/wsbm.1469] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 10/01/2019] [Accepted: 10/13/2019] [Indexed: 12/20/2022]
Abstract
There has been a spurt in structural neuroimaging studies of the effect of hearing loss on the brain. Specifically, magnetic resonance imaging (MRI) and diffusion tensor imaging (DTI) technologies provide an opportunity to quantify changes in gray and white matter structures at the macroscopic scale. To date, there have been 32 MRI and 23 DTI studies that have analyzed structural differences accruing from pre- or peri-lingual pediatric hearing loss with congenital or early onset etiology and postlingual hearing loss in pre-to-late adolescence. Additionally, there have been 15 prospective clinical structural neuroimaging studies of children and adolescents being evaluated for cochlear implants. The results of the 70 studies are summarized in two figures and three tables. Plastic changes in the brain are seen to be multifocal rather than diffuse, that is, differences are consistent across regions implicated in the hearing, speech and language networks regardless of modes of communication and amplification. Structures in that play an important role in cognition are affected to a lesser extent. A limitation of these studies is the emphasis on volumetric measures and on homogeneous groups of subjects with hearing loss. It is suggested that additional measures of morphometry and connectivity could contribute to a greater understanding of the effect of hearing loss on the brain. Then an interpretation of the observed macroscopic structural differences is given. This is followed by discussion of how structural imaging can be combined with functional imaging to provide biomarkers for longitudinal tracking of amplification. This article is categorized under: Developmental Biology > Developmental Processes in Health and Disease Translational, Genomic, and Systems Medicine > Translational Medicine Laboratory Methods and Technologies > Imaging.
Collapse
Affiliation(s)
- J. Tilak Ratnanather
- Center for Imaging Science, Johns Hopkins University, Baltimore, Maryland
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
18
|
Hruskova B, Trojanova J, Kralikova M, Melichar A, Suchankova S, Bartosova J, Burianova JS, Popelar J, Syka J, Turecek R. Cochlear ablation in neonatal rats disrupts inhibitory transmission in the medial nucleus of the trapezoid body. Neurosci Lett 2019; 699:145-150. [PMID: 30742935 DOI: 10.1016/j.neulet.2019.01.058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 01/11/2019] [Accepted: 01/31/2019] [Indexed: 10/27/2022]
Abstract
Inhibitory circuits in the auditory brainstem undergo multiple postnatal changes that are both activity-dependent and activity-independent. We tested to see if the shift from GABA- to glycinergic transmission, which occurs in the rat medial nucleus of the trapezoid body (MNTB) around the onset of hearing, depends on sound-evoked neuronal activity. We prevented the activity by bilateral cochlear ablations in early postnatal rats and studied ionotropic GABA and glycine receptors in MNTB neurons after hearing onset. The removal of the cochlea decreased responses of GABAA and glycine receptors to exogenous agonists as well as the amplitudes of inhibitory postsynaptic currents. The reduction was accompanied by a decrease in the number of glycine receptor- or vesicular GABA transporter-immunopositive puncta. Furthermore, the ablations markedly affected the switch in presynaptic GABAA to glycine receptors. The increase in the expression of postsynaptic glycine receptors and the shift in inhibitory transmitters were not prevented. The results suggest that inhibitory transmission in the MNTB is subject to multiple developmental signals and support the idea that auditory experience plays a role in the maturation of the brainstem glycinergic circuits.
Collapse
Affiliation(s)
- Bohdana Hruskova
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 14220 Prague 4 - Krc, Czech Republic
| | - Johana Trojanova
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 14220 Prague 4 - Krc, Czech Republic
| | - Michaela Kralikova
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 14220 Prague 4 - Krc, Czech Republic
| | - Adolf Melichar
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 14220 Prague 4 - Krc, Czech Republic
| | - Stepanka Suchankova
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 14220 Prague 4 - Krc, Czech Republic
| | - Jolana Bartosova
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 14220 Prague 4 - Krc, Czech Republic
| | - Jana Svobodova Burianova
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 14220 Prague 4 - Krc, Czech Republic
| | - Jiri Popelar
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 14220 Prague 4 - Krc, Czech Republic
| | - Josef Syka
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 14220 Prague 4 - Krc, Czech Republic
| | - Rostislav Turecek
- Department of Auditory Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 14220 Prague 4 - Krc, Czech Republic.
| |
Collapse
|
19
|
Barone CM, Douma S, Reijntjes DOJ, Browe BM, Köppl C, Klump G, Park TJ, Pyott SJ. Altered cochlear innervation in developing and mature naked and Damaraland mole rats. J Comp Neurol 2019; 527:2302-2316. [PMID: 30861124 PMCID: PMC6767702 DOI: 10.1002/cne.24682] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 03/06/2019] [Accepted: 03/07/2019] [Indexed: 01/04/2023]
Abstract
Compared to many other rodent species, naked mole rats (Heterocephalus glaber) have elevated auditory thresholds, poor frequency selectivity, and limited ability to localize sound. Because the cochlea is responsible for encoding and relaying auditory signals to the brain, we used immunofluorescence and quantitative image analysis to examine cochlear innervation in mature and developing naked mole rats compared to mice (Mus musculus), gerbils (Meriones unguiculatus), and Damaraland mole rats (Fukomys damarensis), another subterranean rodent. In comparison to mice and gerbils, we observed alterations in afferent and efferent innervation as well as their patterns of developmental refinement in naked and Damaraland mole rats. These alterations were, however, not always shared similarly between naked and Damaraland mole rats. Most conspicuously, in both naked and Damaraland mole rats, inner hair cell (IHC) afferent ribbon density was reduced, whereas outer hair cell afferent ribbon density was increased. Naked and Damaraland mole rats also showed reduced lateral and medial efferent terminal density. Developmentally, naked mole rats showed reduced and prolonged postnatal reorganization of afferent and efferent innervation. Damaraland mole rats showed no evidence of postnatal reorganization. Differences in cochlear innervation specifically between the two subterranean rodents and more broadly among rodents provides insight into the cochlear mechanisms that enhance frequency sensitivity and sound localization, maturation of the auditory system, and the evolutionary adaptations occurring in response to subterranean environments.
Collapse
Affiliation(s)
- Catherine M Barone
- Laboratory of Integrative Neuroscience, Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Sytse Douma
- Department of Otorhinolaryngology and Head/Neck Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Daniël O J Reijntjes
- Department of Otorhinolaryngology and Head/Neck Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Brigitte M Browe
- Laboratory of Integrative Neuroscience, Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Christine Köppl
- Cluster of Excellence "Hearing4All", Department of Neuroscience, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany.,Research Center Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Georg Klump
- Cluster of Excellence "Hearing4All", Department of Neuroscience, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany.,Research Center Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Thomas J Park
- Laboratory of Integrative Neuroscience, Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Sonja J Pyott
- Department of Otorhinolaryngology and Head/Neck Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
20
|
Chung Y, Buechel BD, Sunwoo W, Wagner JD, Delgutte B. Neural ITD Sensitivity and Temporal Coding with Cochlear Implants in an Animal Model of Early-Onset Deafness. J Assoc Res Otolaryngol 2019; 20:37-56. [PMID: 30623319 DOI: 10.1007/s10162-018-00708-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 12/17/2018] [Indexed: 01/04/2023] Open
Abstract
Users of cochlear implant (CI) face challenges in everyday situations such as understanding conversations in noise, even with CIs in both ears. These challenges are related to difficulties with tasks that require fine temporal processing such as discrimination of pulse rates or interaural time differences (ITD), a major cue for sound localization. The degradation in temporal processing and ITD sensitivity are especially acute in those who lost hearing in early childhood. Here, we characterized temporal coding and ITD sensitivity of single neurons in a novel animal model of early-onset deafness. Rabbits were deafened as neonates and deprived of auditory stimulation until they reached adult age when single-unit recordings from the auditory midbrain were made chronically using an unanesthetized preparation. The results are compared to measurements from adult-deafened rabbits with normal auditory development to understand the effect of early-onset deafness on neural temporal coding and ITD sensitivity. Neurons in the inferior colliculus (IC) of early-deafened rabbits were less likely to show sustained, excitatory responses to pulse train stimulation and more likely to show suppressive responses compared to neurons in adult-deaf animals. Fewer neurons showed synchronized responses to pulse trains at any rate in the early-deaf group. In addition, fewer neurons showed significant ITD sensitivity in their overall firing rate in the early-deaf group compared to adult-deaf animals. Neural ITD discrimination thresholds in the early-deaf group were poorer than thresholds in adult-deaf group, especially at high pulse rates. The overall degradation in neural ITD sensitivity is consistent with the difficulties encountered by human CI users with early-onset hearing loss. These results lay the groundwork for investigating whether the degradations in temporal coding and ITD sensitivity observed in early-deaf animals can be reversed by appropriate CI stimulation during development.
Collapse
Affiliation(s)
- Yoojin Chung
- Eaton-Peabody Laboratories, Massachusetts Eye & Ear, Boston, MA, 02114, USA.
- Department of Otolaryngology, Harvard Medical School, Boston, MA, 02115, USA.
| | - Brian D Buechel
- Eaton-Peabody Laboratories, Massachusetts Eye & Ear, Boston, MA, 02114, USA
- Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, MA, 02115, USA
| | - Woongsang Sunwoo
- Eaton-Peabody Laboratories, Massachusetts Eye & Ear, Boston, MA, 02114, USA
- Department of Otolaryngology, Harvard Medical School, Boston, MA, 02115, USA
- Department of Otolaryngology, Gachon University Gil Medical Center, Incheon, 405-760, South Korea
| | - Joseph D Wagner
- Eaton-Peabody Laboratories, Massachusetts Eye & Ear, Boston, MA, 02114, USA
| | - Bertrand Delgutte
- Eaton-Peabody Laboratories, Massachusetts Eye & Ear, Boston, MA, 02114, USA
- Department of Otolaryngology, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
21
|
Wang X, Zorio DAR, Schecterson L, Lu Y, Wang Y. Postsynaptic FMRP Regulates Synaptogenesis In Vivo in the Developing Cochlear Nucleus. J Neurosci 2018; 38:6445-6460. [PMID: 29950504 PMCID: PMC6052239 DOI: 10.1523/jneurosci.0665-18.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/18/2018] [Accepted: 06/20/2018] [Indexed: 12/29/2022] Open
Abstract
A global loss of the fragile X mental retardation protein (FMRP; encoded by the Fmr1 gene) leads to sensory dysfunction and intellectual disabilities. One underlying mechanism of these phenotypes is structural and functional deficits in synapses. Here, we determined the autonomous function of postsynaptic FMRP in circuit formation, synaptogenesis, and synaptic maturation. In normal cochlea nucleus, presynaptic auditory axons form large axosomatic endbulb synapses on cell bodies of postsynaptic bushy neurons. In ovo electroporation of drug-inducible Fmr1-shRNA constructs produced a mosaicism of FMRP expression in chicken (either sex) bushy neurons, leading to reduced FMRP levels in transfected, but not neighboring nontransfected, neurons. Structural analyses revealed that postsynaptic FMRP reduction led to smaller size and abnormal morphology of individual presynaptic endbulbs at both early and later developmental stages. We further examined whether FMRP reduction affects dendritic development, as a potential mechanism underlying defective endbulb formation. Normally, chicken bushy neurons grow extensive dendrites at early stages and retract these dendrites when endbulbs begin to form. Neurons transfected with Fmr1 shRNA exhibited a remarkable delay in branch retraction, failing to provide necessary somatic surface for timely formation and growth of large endbulbs. Patch-clamp recording verified functional consequences of dendritic and synaptic deficits on neurotransmission, showing smaller amplitudes and slower kinetics of spontaneous and evoked EPSCs. Together, these data demonstrate that proper levels of postsynaptic FMRP are required for timely maturation of somatodendritic morphology, a delay of which may affect synaptogenesis and thus contribute to long-lasting deficits of excitatory synapses.SIGNIFICANCE STATEMENT Fragile X mental retardation protein (FMRP) regulates a large variety of neuronal activities. A global loss of FMRP affects neural circuit development and synaptic function, leading to fragile X syndrome (FXS). Using temporally and spatially controlled genetic manipulations, this study provides the first in vivo report that autonomous FMRP regulates multiple stages of dendritic development, and that selective reduction of postsynaptic FMRP leads to abnormal development of excitatory presynaptic terminals and compromised neurotransmission. These observations demonstrate secondary influence of developmentally transient deficits in neuronal morphology and connectivity to the development of long-lasting synaptic pathology in FXS.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Department of Biomedical Science, Program in Neuroscience, Florida State University College of Medicine, Tallahassee, Florida 32306
| | - Diego A R Zorio
- Department of Biomedical Science, Program in Neuroscience, Florida State University College of Medicine, Tallahassee, Florida 32306
| | - Leslayann Schecterson
- Department of Otolaryngology, Bloedel Hearing Research Center, University of Washington, Seattle, Washington 98195, and
| | - Yong Lu
- Department of Anatomy and Neurobiology, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio 44272
| | - Yuan Wang
- Department of Biomedical Science, Program in Neuroscience, Florida State University College of Medicine, Tallahassee, Florida 32306,
| |
Collapse
|
22
|
Changes in Properties of Auditory Nerve Synapses following Conductive Hearing Loss. J Neurosci 2017; 37:323-332. [PMID: 28077712 DOI: 10.1523/jneurosci.0523-16.2016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 11/10/2016] [Accepted: 11/19/2016] [Indexed: 02/08/2023] Open
Abstract
Auditory activity plays an important role in the development of the auditory system. Decreased activity can result from conductive hearing loss (CHL) associated with otitis media, which may lead to long-term perceptual deficits. The effects of CHL have been mainly studied at later stages of the auditory pathway, but early stages remain less examined. However, changes in early stages could be important because they would affect how information about sounds is conveyed to higher-order areas for further processing and localization. We examined the effects of CHL at auditory nerve synapses onto bushy cells in the mouse anteroventral cochlear nucleus following occlusion of the ear canal. These synapses, called endbulbs of Held, normally show strong depression in voltage-clamp recordings in brain slices. After 1 week of CHL, endbulbs showed even greater depression, reflecting higher release probability. We observed no differences in quantal size between control and occluded mice. We confirmed these observations using mean-variance analysis and the integration method, which also revealed that the number of release sites decreased after occlusion. Consistent with this, synaptic puncta immunopositive for VGLUT1 decreased in area after occlusion. The level of depression and number of release sites both showed recovery after returning to normal conditions. Finally, bushy cells fired fewer action potentials in response to evoked synaptic activity after occlusion, likely because of increased depression and decreased input resistance. These effects appear to reflect a homeostatic, adaptive response of auditory nerve synapses to reduced activity. These effects may have important implications for perceptual changes following CHL. SIGNIFICANCE STATEMENT Normal hearing is important to everyday life, but abnormal auditory experience during development can lead to processing disorders. For example, otitis media reduces sound to the ear, which can cause long-lasting deficits in language skills and verbal production, but the location of the problem is unknown. Here, we show that occluding the ear causes synapses at the very first stage of the auditory pathway to modify their properties, by decreasing in size and increasing the likelihood of releasing neurotransmitter. This causes synapses to deplete faster, which reduces fidelity at central targets of the auditory nerve, which could affect perception. Temporary hearing loss could cause similar changes at later stages of the auditory pathway, which could contribute to disorders in behavior.
Collapse
|
23
|
Guo Y, Zhang P, Sheng Q, Zhao S, Hackett TA. lncRNA expression in the auditory forebrain during postnatal development. Gene 2016; 593:201-216. [PMID: 27544636 PMCID: PMC5034298 DOI: 10.1016/j.gene.2016.08.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 06/27/2016] [Accepted: 08/15/2016] [Indexed: 12/30/2022]
Abstract
The biological processes governing brain development and maturation depend on complex patterns of gene and protein expression, which can be influenced by many factors. One of the most overlooked is the long noncoding class of RNAs (lncRNAs), which are known to play important regulatory roles in an array of biological processes. Little is known about the distribution of lncRNAs in the sensory systems of the brain, and how lncRNAs interact with other mechanisms to guide the development of these systems. In this study, we profiled lncRNA expression in the mouse auditory forebrain during postnatal development at time points before and after the onset of hearing (P7, P14, P21, adult). First, we generated lncRNA profiles of the primary auditory cortex (A1) and medial geniculate body (MG) at each age. Then, we determined the differential patterns of expression by brain region and age. These analyses revealed that the lncRNA expression profile was distinct between both brain regions and between each postnatal age, indicating spatial and temporal specificity during maturation of the auditory forebrain. Next, we explored potential interactions between functionally-related lncRNAs, protein coding RNAs (pcRNAs), and associated proteins. The maturational trajectories (P7 to adult) of many lncRNA - pcRNA pairs were highly correlated, and predictive analyses revealed that lncRNA-protein interactions tended to be strong. A user-friendly database was constructed to facilitate inspection of the expression levels and maturational trajectories for any lncRNA or pcRNA in the database. Overall, this study provides an in-depth summary of lncRNA expression in the developing auditory forebrain and a broad-based foundation for future exploration of lncRNA function during brain development.
Collapse
Affiliation(s)
- Yan Guo
- Dept. of Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - Pan Zhang
- Dept. of Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - Quanhu Sheng
- Dept. of Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - Shilin Zhao
- Dept. of Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | - Troy A Hackett
- Dept. of Hearing and Speech Sciences, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
24
|
Vollmer M, Beitel RE, Schreiner CE, Leake PA. Passive stimulation and behavioral training differentially transform temporal processing in the inferior colliculus and primary auditory cortex. J Neurophysiol 2016; 117:47-64. [PMID: 27733594 DOI: 10.1152/jn.00392.2016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 10/05/2016] [Indexed: 11/22/2022] Open
Abstract
In profoundly deaf cats, behavioral training with intracochlear electric stimulation (ICES) can improve temporal processing in the primary auditory cortex (AI). To investigate whether similar effects are manifest in the auditory midbrain, ICES was initiated in neonatally deafened cats either during development after short durations of deafness (8 wk of age) or in adulthood after long durations of deafness (≥3.5 yr). All of these animals received behaviorally meaningless, "passive" ICES. Some animals also received behavioral training with ICES. Two long-deaf cats received no ICES prior to acute electrophysiological recording. After several months of passive ICES and behavioral training, animals were anesthetized, and neuronal responses to pulse trains of increasing rates were recorded in the central (ICC) and external (ICX) nuclei of the inferior colliculus. Neuronal temporal response patterns (repetition rate coding, minimum latencies, response precision) were compared with results from recordings made in the AI of the same animals (Beitel RE, Vollmer M, Raggio MW, Schreiner CE. J Neurophysiol 106: 944-959, 2011; Vollmer M, Beitel RE. J Neurophysiol 106: 2423-2436, 2011). Passive ICES in long-deaf cats remediated severely degraded temporal processing in the ICC and had no effects in the ICX. In contrast to observations in the AI, behaviorally relevant ICES had no effects on temporal processing in the ICC or ICX, with the single exception of shorter latencies in the ICC in short-deaf cats. The results suggest that independent of deafness duration passive stimulation and behavioral training differentially transform temporal processing in auditory midbrain and cortex, and primary auditory cortex emerges as a pivotal site for behaviorally driven neuronal temporal plasticity in the deaf cat. NEW & NOTEWORTHY Behaviorally relevant vs. passive electric stimulation of the auditory nerve differentially affects neuronal temporal processing in the central nucleus of the inferior colliculus (ICC) and the primary auditory cortex (AI) in profoundly short-deaf and long-deaf cats. Temporal plasticity in the ICC depends on a critical amount of electric stimulation, independent of its behavioral relevance. In contrast, the AI emerges as a pivotal site for behaviorally driven neuronal temporal plasticity in the deaf auditory system.
Collapse
Affiliation(s)
- Maike Vollmer
- Comprehensive Hearing Center, University Hospital Wuerzburg, Wuerzburg, Germany;
| | - Ralph E Beitel
- Coleman Memorial Laboratory, Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, California
| | - Christoph E Schreiner
- Center for Integrative Neuroscience, Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, California; and
| | - Patricia A Leake
- Epstein Laboratory, Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, California
| |
Collapse
|
25
|
Corneal-Reflection Eye-Tracking Technique for the Assessment of Horizontal Sound Localization Accuracy from 6 Months of Age. Ear Hear 2016; 37:e104-18. [DOI: 10.1097/aud.0000000000000235] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
26
|
Hackett TA, Guo Y, Clause A, Hackett NJ, Garbett K, Zhang P, Polley DB, Mirnics K. Transcriptional maturation of the mouse auditory forebrain. BMC Genomics 2015; 16:606. [PMID: 26271746 PMCID: PMC4536593 DOI: 10.1186/s12864-015-1709-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 06/01/2015] [Indexed: 02/07/2023] Open
Abstract
Background The maturation of the brain involves the coordinated expression of thousands of genes, proteins and regulatory elements over time. In sensory pathways, gene expression profiles are modified by age and sensory experience in a manner that differs between brain regions and cell types. In the auditory system of altricial animals, neuronal activity increases markedly after the opening of the ear canals, initiating events that culminate in the maturation of auditory circuitry in the brain. This window provides a unique opportunity to study how gene expression patterns are modified by the onset of sensory experience through maturity. As a tool for capturing these features, next-generation sequencing of total RNA (RNAseq) has tremendous utility, because the entire transcriptome can be screened to index expression of any gene. To date, whole transcriptome profiles have not been generated for any central auditory structure in any species at any age. In the present study, RNAseq was used to profile two regions of the mouse auditory forebrain (A1, primary auditory cortex; MG, medial geniculate) at key stages of postnatal development (P7, P14, P21, adult) before and after the onset of hearing (~P12). Hierarchical clustering, differential expression, and functional geneset enrichment analyses (GSEA) were used to profile the expression patterns of all genes. Selected genesets related to neurotransmission, developmental plasticity, critical periods and brain structure were highlighted. An accessible repository of the entire dataset was also constructed that permits extraction and screening of all data from the global through single-gene levels. To our knowledge, this is the first whole transcriptome sequencing study of the forebrain of any mammalian sensory system. Although the data are most relevant for the auditory system, they are generally applicable to forebrain structures in the visual and somatosensory systems, as well. Results The main findings were: (1) Global gene expression patterns were tightly clustered by postnatal age and brain region; (2) comparing A1 and MG, the total numbers of differentially expressed genes were comparable from P7 to P21, then dropped to nearly half by adulthood; (3) comparing successive age groups, the greatest numbers of differentially expressed genes were found between P7 and P14 in both regions, followed by a steady decline in numbers with age; (4) maturational trajectories in expression levels varied at the single gene level (increasing, decreasing, static, other); (5) between regions, the profiles of single genes were often asymmetric; (6) GSEA revealed that genesets related to neural activity and plasticity were typically upregulated from P7 to adult, while those related to structure tended to be downregulated; (7) GSEA and pathways analysis of selected functional networks were not predictive of expression patterns in the auditory forebrain for all genes, reflecting regional specificity at the single gene level. Conclusions Gene expression in the auditory forebrain during postnatal development is in constant flux and becomes increasingly stable with age. Maturational changes are evident at the global through single gene levels. Transcriptome profiles in A1 and MG are distinct at all ages, and differ from other brain regions. The database generated by this study provides a rich foundation for the identification of novel developmental biomarkers, functional gene pathways, and targeted studies of postnatal maturation in the auditory forebrain. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1709-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Troy A Hackett
- Department of Hearing and Speech Sciences, Vanderbilt University School of Medicine, Nashville, TN, USA. .,Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN, 37232, USA.
| | - Yan Guo
- Department of Cancer Biology, Vanderbilt University, Nashville, TN, USA.
| | - Amanda Clause
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear Infirmary, Department of Otology and Laryngology, Harvard Medical School, Boston, MA, USA.
| | | | | | - Pan Zhang
- Department of Cancer Biology, Vanderbilt University, Nashville, TN, USA.
| | - Daniel B Polley
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear Infirmary, Department of Otology and Laryngology, Harvard Medical School, Boston, MA, USA.
| | - Karoly Mirnics
- Department of Psychiatry, Vanderbilt University, Nashville, TN, USA. .,Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN, 37235, USA. .,Department of Psychiatry, University of Szeged, 6725, Szeged, Hungary. .,Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
27
|
Asp F, Mäki-Torkko E, Karltorp E, Harder H, Hergils L, Eskilsson G, Stenfelt S. A longitudinal study of the bilateral benefit in children with bilateral cochlear implants. Int J Audiol 2014; 54:77-88. [DOI: 10.3109/14992027.2014.973536] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
28
|
Postnatal expression of neurotrophic factors accessible to spiral ganglion neurons in the auditory system of adult hearing and deafened rats. J Neurosci 2014; 34:13110-26. [PMID: 25253857 DOI: 10.1523/jneurosci.1014-14.2014] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Spiral ganglion neurons (SGNs) receive input from cochlear hair cells and project from the cochlea to the cochlear nucleus. After destruction of hair cells with aminoglycoside antibiotics or noise, SGNs gradually die. It has been assumed that SGN death is attributable to loss of neurotrophic factors (NTFs) derived from hair cells or supporting cells in the organ of Corti (OC). We used quantitative PCR (qPCR) to assay NTF expression-neurotrophin-3 (NT-3), BDNF, GDNF, neurturin, artemin, and CNTF-in the OC and cochlear nucleus at various ages from postnatal day 0 (P0) to P90 in control hearing and neonatally deafened rats. NT-3, neurturin, and CNTF were most abundant in the postnatal hearing OC; CNTF and neurturin most abundant in the cochlear nucleus. In the OC, NT-3 and CNTF showed a postnatal increase in expression approximately concomitant with hearing onset. In rats deafened by daily kanamycin injections (from P8 to P16), surviving inner hair cells were evident at P16 but absent by P19, with most postsynaptic boutons lost before P16. NT-3 and CNTF, which normally increase postnatally, had significantly reduced expression in the OC of deafened rats, although CNTF was expressed throughout the time that SGNs were dying. In contrast, neurturin expression was constant, unaffected by deafening or by age. CNTF and neurturin expression in the cochlear nucleus was unaffected by deafening or age. Thus, NTFs other than NT-3 are available to SGNs even as they are dying after deafening, apparently conflicting with the hypothesis that SGN death is attributable to lack of NTFs.
Collapse
|
29
|
Canlon B. Progress in hearing research 2014. Hear Res 2014; 311:1-2. [PMID: 25151010 DOI: 10.1016/j.heares.2014.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 02/18/2014] [Indexed: 11/19/2022]
Affiliation(s)
- Barbara Canlon
- Karolinska Institutet, Department of Physiology and Pharmacology, von Eulers vag 8, 171 77 Stockholm, Sweden.
| |
Collapse
|
30
|
Rubio ME, Fukazawa Y, Kamasawa N, Clarkson C, Molnár E, Shigemoto R. Target- and input-dependent organization of AMPA and NMDA receptors in synaptic connections of the cochlear nucleus. J Comp Neurol 2014; 522:4023-42. [PMID: 25041792 DOI: 10.1002/cne.23654] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Revised: 07/07/2014] [Accepted: 07/17/2014] [Indexed: 12/12/2022]
Abstract
We examined the synaptic structure, quantity, and distribution of α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA)- and N-methyl-D-aspartate (NMDA)-type glutamate receptors (AMPARs and NMDARs, respectively) in rat cochlear nuclei by a highly sensitive freeze-fracture replica labeling technique. Four excitatory synapses formed by two distinct inputs, auditory nerve (AN) and parallel fibers (PF), on different cell types were analyzed. These excitatory synapse types included AN synapses on bushy cells (AN-BC synapses) and fusiform cells (AN-FC synapses) and PF synapses on FC (PF-FC synapses) and cartwheel cell spines (PF-CwC synapses). Immunogold labeling revealed differences in synaptic structure as well as AMPAR and NMDAR number and/or density in both AN and PF synapses, indicating a target-dependent organization. The immunogold receptor labeling also identified differences in the synaptic organization of FCs based on AN or PF connections, indicating an input-dependent organization in FCs. Among the four excitatory synapse types, the AN-BC synapses were the smallest and had the most densely packed intramembrane particles (IMPs), whereas the PF-CwC synapses were the largest and had sparsely packed IMPs. All four synapse types showed positive correlations between the IMP-cluster area and the AMPAR number, indicating a common intrasynapse-type relationship for glutamatergic synapses. Immunogold particles for AMPARs were distributed over the entire area of individual AN synapses; PF synapses often showed synaptic areas devoid of labeling. The gold-labeling for NMDARs occurred in a mosaic fashion, with less positive correlations between the IMP-cluster area and the NMDAR number. Our observations reveal target- and input-dependent features in the structure, number, and organization of AMPARs and NMDARs in AN and PF synapses.
Collapse
Affiliation(s)
- María E Rubio
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Neurobiology, University of Pittsburgh, PA, USA
| | | | | | | | | | | |
Collapse
|
31
|
Ulanet PG, Carson CM, Mellon NK, Niparko JK, Ouellette M. Correlation of neurocognitive processing subtypes with language performance in young children with cochlear implants. Cochlear Implants Int 2014; 15:230-40. [PMID: 24840711 DOI: 10.1179/1754762814y.0000000077] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES Test data were used to explore the neurocognitive processing of a group of children with cochlear implants (CIs) whose language development is below expectations. METHODS This cross-sectional study examines the relationship between neurocognitive processing, as assessed by the Kaufman Assessment Battery for Children-Second Edition, and verbal language standard scores, assessed using either the Comprehensive Assessment of Spoken Language or the Clinical Evaluation of Language Fundamentals in 22 school-age children with CIs. Processing scores of CI recipients with language scores below expectations were compared to those of children meeting or exceeding language expectations. Multiple linear regression estimated the associations of simultaneous and sequential processing with language scores. RESULTS Though simultaneous processing scores between the two groups were similar, the mean sequential processing score (91.2) in the below expectations group (n = 13) was significantly lower (P = 0.002) than that of children (n = 9) meeting expectations (110.8). After adjusting for age at implantation, a 10-point higher sequential processing score was associated with a 7.4 higher language score (P = 0.027). DISCUSSION Simultaneous processing capacity was at least within the average range of cognitive performance, and was not associated with language performance in children with CIs. Conversely, reduced sequential processing capacity was significantly associated with lower language scores. CONCLUSION Neurocognitive skills, specifically cognitive sequencing, serial ordering, and auditory-verbal memory may be targets for therapeutic intervention. Intensive cognitive and educational habilitation and in milieu intervention may improve language learning in children with CIs.
Collapse
|
32
|
Morphological and physiological development of auditory synapses. Hear Res 2014; 311:3-16. [PMID: 24508369 DOI: 10.1016/j.heares.2014.01.007] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Revised: 01/13/2014] [Accepted: 01/20/2014] [Indexed: 02/07/2023]
Abstract
Acoustic communication requires gathering, transforming, and interpreting diverse sound cues. To achieve this, all the spatial and temporal features of complex sound stimuli must be captured in the firing patterns of the primary sensory neurons and then accurately transmitted along auditory pathways for additional processing. The mammalian auditory system relies on several synapses with unique properties in order to meet this task: the auditory ribbon synapses, the endbulb of Held, and the calyx of Held. Each of these synapses develops morphological and electrophysiological characteristics that enable the remarkably precise signal transmission necessary for conveying the miniscule differences in timing that underly sound localization. In this article, we review the current knowledge of how these synapses develop and mature to acquire the specialized features necessary for the sense of hearing.
Collapse
|
33
|
Mendoza Schulz A, Jing Z, Sánchez Caro JM, Wetzel F, Dresbach T, Strenzke N, Wichmann C, Moser T. Bassoon-disruption slows vesicle replenishment and induces homeostatic plasticity at a CNS synapse. EMBO J 2014; 33:512-27. [PMID: 24442636 DOI: 10.1002/embj.201385887] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Endbulb of Held terminals of auditory nerve fibers (ANF) transmit auditory information at hundreds per second to bushy cells (BCs) in the anteroventral cochlear nucleus (AVCN). Here, we studied the structure and function of endbulb synapses in mice that lack the presynaptic scaffold bassoon and exhibit reduced ANF input into the AVCN. Endbulb terminals and active zones were normal in number and vesicle complement. Postsynaptic densities, quantal size and vesicular release probability were increased while vesicle replenishment and the standing pool of readily releasable vesicles were reduced. These opposing effects canceled each other out for the first evoked EPSC, which showed unaltered amplitude. We propose that ANF activity deprivation drives homeostatic plasticity in the AVCN involving synaptic upscaling and increased intrinsic BC excitability. In vivo recordings from individual mutant BCs demonstrated a slightly improved response at sound onset compared to ANF, likely reflecting the combined effects of ANF convergence and homeostatic plasticity. Further, we conclude that bassoon promotes vesicular replenishment and, consequently, a large standing pool of readily releasable synaptic vesicles at the endbulb synapse.
Collapse
Affiliation(s)
- Alejandro Mendoza Schulz
- InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Characterization of human auditory brainstem circuits by calcium-binding protein immunohistochemistry. Neuroscience 2014; 258:318-31. [DOI: 10.1016/j.neuroscience.2013.11.035] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 11/05/2013] [Accepted: 11/19/2013] [Indexed: 11/18/2022]
|
35
|
Butler BE, Lomber SG. Functional and structural changes throughout the auditory system following congenital and early-onset deafness: implications for hearing restoration. Front Syst Neurosci 2013; 7:92. [PMID: 24324409 PMCID: PMC3840613 DOI: 10.3389/fnsys.2013.00092] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 11/03/2013] [Indexed: 11/23/2022] Open
Abstract
The absence of auditory input, particularly during development, causes widespread changes in the structure and function of the auditory system, extending from peripheral structures into auditory cortex. In humans, the consequences of these changes are far-reaching and often include detriments to language acquisition, and associated psychosocial issues. Much of what is currently known about the nature of deafness-related changes to auditory structures comes from studies of congenitally deaf or early-deafened animal models. Fortunately, the mammalian auditory system shows a high degree of preservation among species, allowing for generalization from these models to the human auditory system. This review begins with a comparison of common methods used to obtain deaf animal models, highlighting the specific advantages and anatomical consequences of each. Some consideration is also given to the effectiveness of methods used to measure hearing loss during and following deafening procedures. The structural and functional consequences of congenital and early-onset deafness have been examined across a variety of mammals. This review attempts to summarize these changes, which often involve alteration of hair cells and supporting cells in the cochleae, and anatomical and physiological changes that extend through subcortical structures and into cortex. The nature of these changes is discussed, and the impacts to neural processing are addressed. Finally, long-term changes in cortical structures are discussed, with a focus on the presence or absence of cross-modal plasticity. In addition to being of interest to our understanding of multisensory processing, these changes also have important implications for the use of assistive devices such as cochlear implants.
Collapse
Affiliation(s)
- Blake E. Butler
- Cerebral Systems Laboratory, Department of Physiology and Pharmacology, Brain and Mind Institute, University of Western OntarioLondon, ON, Canada
| | - Stephen G. Lomber
- Cerebral Systems Laboratory, Department of Physiology and Pharmacology and Department of Psychology, National Centre for Audiology, Brain and Mind Institute, University of Western OntarioLondon, ON, Canada
| |
Collapse
|
36
|
Abstract
In the developing nervous system, spontaneous neuronal activity arises independently of experience or any environmental input. This activity may play a major role in axonal pathfinding, refinement of topographic maps, dendritic morphogenesis, and the segregation of axonal terminal arbors. In the auditory system, endogenously released ATP in the cochlea activates inner hair cells to trigger bursts of action potentials (APs), which are transferred to the central auditory system. Here we show the modulatory role of purinergic signaling beyond the cochlea, i.e., the developmentally regulated and cell-type-specific depolarizing effects on auditory brainstem neurons of Mongolian gerbil. We assessed the effects of P2X receptors (P2XRs) on neuronal excitability from prehearing to early stages of auditory signal processing. Our results demonstrate that in neurons expressing P2XRs, extracellular ATP can evoke APs in sync with Ca(2+) signals. In cochlear nucleus (CN) bushy cells, ATP increases spontaneous and also acoustically evoked activity in vivo, but these effects diminish with maturity. Moreover, ATP not only augmented glutamate-driven firing, but it also evoked APs in the absence of glutamatergic transmission. In vivo recordings also revealed that endogenously released ATP in the CN contributes to neuronal firing activity by facilitating AP generation and prolonging AP duration. Given the enhancing effect of ATP on AP firing and confinement of P2XRs to certain auditory brainstem nuclei, and to distinct neurons within these nuclei, it is conceivable that purinergic signaling plays a specific role in the development of neuronal brainstem circuits.
Collapse
|
37
|
Tang Y, Christensen-Dalsgaard J, Carr CE. Organization of the auditory brainstem in a lizard, Gekko gecko. I. Auditory nerve, cochlear nuclei, and superior olivary nuclei. J Comp Neurol 2012; 520:1784-99. [PMID: 22120438 PMCID: PMC4300985 DOI: 10.1002/cne.23013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We used tract tracing to reveal the connections of the auditory brainstem in the Tokay gecko (Gekko gecko). The auditory nerve has two divisions, a rostroventrally directed projection of mid- to high best-frequency fibers to the nucleus angularis (NA) and a more dorsal and caudal projection of low to middle best-frequency fibers that bifurcate to project to both the NA and the nucleus magnocellularis (NM). The projection to NM formed large somatic terminals and bouton terminals. NM projected bilaterally to the second-order nucleus laminaris (NL), such that the ipsilateral projection innervated the dorsal NL neuropil, whereas the contralateral projection crossed the midline and innervated the ventral dendrites of NL neurons. Neurons in NL were generally bitufted, with dorsoventrally oriented dendrites. NL projected to the contralateral torus semicircularis and to the contralateral ventral superior olive (SOv). NA projected to ipsilateral dorsal superior olive (SOd), sent a major projection to the contralateral SOv, and projected to torus semicircularis. The SOd projected to the contralateral SOv, which projected back to the ipsilateral NM, NL, and NA. These results suggest homologous patterns of auditory connections in lizards and archosaurs but also different processing of low- and high-frequency information in the brainstem.
Collapse
Affiliation(s)
- Yezhong Tang
- Chengdu Institute of Biology, CAS, Chengdu, 610041 People's Republic of China.
| | | | | |
Collapse
|
38
|
Abstract
Long-term binaural auditory deprivation is associated with poorer speech recognition outcomes after cochlear implantation, even for postlingual hearing loss. It is, however, unknown to what extent the outcomes of implantation are related to the peripheral changes occurring monaurally or to changes at a higher level in the auditory system related to binaural deafness. This retrospective study aimed to unravel peripheral and central contributions to cochlear implantation outcomes by comparing outcomes obtained in individual ears for adults with long-term monaural auditory deprivation (i.e. unilateral use of hearing aid) who received bilateral cochlear implants. Results showed that similar outcomes can be obtained with the implant placed in the auditory-deprived or in the aided ear. This suggests that the peripheral changes related to monaural auditory deprivation have little effect on outcomes of cochlear implantation.
Collapse
|