1
|
Maximov K, Kanold PO. Aging reduces excitatory bandwidth, alters spectral tuning curve diversity, and reduces sideband inhibition in L2/3 of primary auditory cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.02.646797. [PMID: 40236140 PMCID: PMC11996523 DOI: 10.1101/2025.04.02.646797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Presbycusis, or age-related hearing loss, is caused by changes in both the peripheral and the central auditory system. Many of the peripheral structures that degrade with age have been identified and characterized, but there is still a dearth of information pertaining to what changes occur in the aging central auditory pathway that are independent of peripheral degradation. The primary auditory cortex (A1) of aging mice shows reduced suppressive responses and reduced diversity of temporal responses suggesting alteration of inhibitory processing. To gain a better understanding of how tuning features of the auditory cortex change with age, we performed in vivo 2-photon Ca 2+ imaging on L2/3 of the auditory cortex of both adult (n=14, 11-24 weeks old) and aging (n=12, 12-17 months old) mice that retain peripheral hearing in old age. To reveal inhibitory inputs to L2/3 neurons we characterized spectral receptive fields with pure tones and two tone complexes. We find that in contrast to adult mice, L2/3 excitatory neurons from aging mice showed fewer distinct categories of spectral receptive fields, though in a subset of FRA types, we found increased diversity. We also noted a decrease in excitatory bandwidth with age among broadly tuned neurons, but that sideband inhibition became weaker across all FRA types due to a reduced amplitude in inhibitory responses. These results suggest that aging causes changes in circuit organization leading to more homogenous spectrotemporal receptive fields and that the lack of response diversity contributes to a decreased encoding capacity observed in aging A1.
Collapse
|
2
|
Xu Z, Xue B, Kao JPY, Kanold PO. Sex-Specific Age-Related Changes in Excitatory and Inhibitory Intracortical Circuits in Mouse Primary Auditory Cortex. eNeuro 2025; 12:ENEURO.0378-24.2024. [PMID: 39626952 PMCID: PMC11826992 DOI: 10.1523/eneuro.0378-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/11/2024] [Indexed: 02/08/2025] Open
Abstract
A common impairment in aging is age-related hearing loss (presbycusis), which manifests as impaired spectrotemporal processing. Presbycusis can be caused by a dysfunction of the peripheral and central auditory system, and these dysfunctions might differ between the sexes. To date, the circuit mechanisms in the central nervous system responsible for age-related auditory dysfunction remain mostly unknown. In the auditory cortex (ACtx), aging is accompanied by alteration in normal inhibitory (GABA) neurotransmission and changes in excitatory (NMDA and AMPA) synapses, but which circuits are affected has been unclear. Here we investigated how auditory cortical microcircuits change with age and if sex-dependent differences existed. We performed laser-scanning photostimulation (LSPS) combined with whole-cell patch-clamp recordings from layer (L) 2/3 cells in the primary auditory cortex (A1) in young adult (2-3 months) and aged (older than 18 months) male and female CBA/CaJ mice which have normal peripheral hearing. We found that L2/3 cells in aged male animals display functional hypoconnectivity of inhibitory circuits originating from L2/3 and L4. Compared with cells from young adult mice, cells from aged male mice have weaker excitatory connections from L2/3. We also observed an increased diversity of excitatory and inhibitory inputs. These results suggest a sex-specific reduction and diversification in excitatory and inhibitory intralaminar cortical circuits in aged mice compared with young adult animals. We speculate that these unbalanced changes in cortical circuits contribute to the functional manifestations of age-related hearing loss in both males and females.
Collapse
Affiliation(s)
- Zheng Xu
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 20215
| | - Binghan Xue
- Department of Biology, University of Maryland, College Park, Maryland 20742
| | - Joseph P Y Kao
- Center for Biomedical Engineering and Technology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Patrick O Kanold
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 20215
- Department of Biology, University of Maryland, College Park, Maryland 20742
| |
Collapse
|
3
|
Wake N, Shiramatsu TI, Takahashi H. Map plasticity following noise exposure in auditory cortex of rats: implications for disentangling neural correlates of tinnitus and hyperacusis. Front Neurosci 2024; 18:1385942. [PMID: 38881748 PMCID: PMC11176560 DOI: 10.3389/fnins.2024.1385942] [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: 02/14/2024] [Accepted: 05/16/2024] [Indexed: 06/18/2024] Open
Abstract
Introduction Both tinnitus and hyperacusis, likely triggered by hearing loss, can be attributed to maladaptive plasticity in auditory perception. However, owing to their co-occurrence, disentangling their neural mechanisms proves difficult. We hypothesized that the neural correlates of tinnitus are associated with neural activities triggered by low-intensity tones, while hyperacusis is linked to responses to moderate- and high-intensity tones. Methods To test these hypotheses, we conducted behavioral and electrophysiological experiments in rats 2 to 8 days after traumatic tone exposure. Results In the behavioral experiments, prepulse and gap inhibition tended to exhibit different frequency characteristics (although not reaching sufficient statistical levels), suggesting that exposure to traumatic tones led to acute symptoms of hyperacusis and tinnitus at different frequency ranges. When examining the auditory cortex at the thalamocortical recipient layer, we observed that tinnitus symptoms correlated with a disorganized tonotopic map, typically characterized by responses to low-intensity tones. Neural correlates of hyperacusis were found in the cortical recruitment function at the multi-unit activity (MUA) level, but not at the local field potential (LFP) level, in response to moderate- and high-intensity tones. This shift from LFP to MUA was associated with a loss of monotonicity, suggesting a crucial role for inhibitory synapses. Discussion Thus, in acute symptoms of traumatic tone exposure, our experiments successfully disentangled the neural correlates of tinnitus and hyperacusis at the thalamocortical recipient layer of the auditory cortex. They also suggested that tinnitus is linked to central noise, whereas hyperacusis is associated with aberrant gain control. Further interactions between animal experiments and clinical studies will offer insights into neural mechanisms, diagnosis and treatments of tinnitus and hyperacusis, specifically in terms of long-term plasticity of chronic symptoms.
Collapse
Affiliation(s)
- Naoki Wake
- Department of Mechano-Informatics, Graduate School of Information Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Tomoyo I Shiramatsu
- Department of Mechano-Informatics, Graduate School of Information Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Hirokazu Takahashi
- Department of Mechano-Informatics, Graduate School of Information Science and Technology, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
4
|
Parameshwarappa V, Siponen MI, Watabe I, Karkaba A, Galazyuk A, Noreña AJ. Noise-induced hearing loss alters potassium-chloride cotransporter KCC2 and GABA inhibition in the auditory centers. Sci Rep 2024; 14:10689. [PMID: 38724641 PMCID: PMC11082187 DOI: 10.1038/s41598-024-60858-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
Homeostatic plasticity, the ability of neurons to maintain their averaged activity constant around a set point value, is thought to account for the central hyperactivity after hearing loss. Here, we investigated the putative role of GABAergic neurotransmission in this mechanism after a noise-induced hearing loss larger than 50 dB in high frequencies in guinea pigs. The effect of GABAergic inhibition is linked to the normal functioning of K + -Cl- co-transporter isoform 2 (KCC2) which maintains a low intracellular concentration of chloride. The expression of membrane KCC2 were investigated before and after noise trauma in the ventral and dorsal cochlear nucleus (VCN and DCN, respectively) and in the inferior colliculus (IC). Moreover, the effect of gabazine (GBZ), a GABA antagonist, was also studied on the neural activity in IC. We show that KCC2 is downregulated in VCN, DCN and IC 3 days after noise trauma, and in DCN and IC 30 days after the trauma. As expected, GBZ application in the IC of control animals resulted in an increase of spontaneous and stimulus-evoked activity. In the noise exposed animals, on the other hand, GBZ application decreased the stimulus-evoked activity in IC neurons. The functional implications of these central changes are discussed.
Collapse
Affiliation(s)
- V Parameshwarappa
- Laboratory of Cognitive Neurosciences, Centre National de la Recherche Scientifique, Aix-Marseille University, 3 Place Victor Hugo, 13003, Marseille, France
| | - M I Siponen
- Laboratory of Cognitive Neurosciences, Centre National de la Recherche Scientifique, Aix-Marseille University, 3 Place Victor Hugo, 13003, Marseille, France
| | - I Watabe
- Laboratory of Cognitive Neurosciences, Centre National de la Recherche Scientifique, Aix-Marseille University, 3 Place Victor Hugo, 13003, Marseille, France
| | - A Karkaba
- Laboratory of Cognitive Neurosciences, Centre National de la Recherche Scientifique, Aix-Marseille University, 3 Place Victor Hugo, 13003, Marseille, France
| | - A Galazyuk
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH, USA
| | - A J Noreña
- Laboratory of Cognitive Neurosciences, Centre National de la Recherche Scientifique, Aix-Marseille University, 3 Place Victor Hugo, 13003, Marseille, France.
| |
Collapse
|
5
|
McClaskey CM. Neural hyperactivity and altered envelope encoding in the central auditory system: Changes with advanced age and hearing loss. Hear Res 2024; 442:108945. [PMID: 38154191 PMCID: PMC10942735 DOI: 10.1016/j.heares.2023.108945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 12/04/2023] [Accepted: 12/22/2023] [Indexed: 12/30/2023]
Abstract
Temporal modulations are ubiquitous features of sound signals that are important for auditory perception. The perception of temporal modulations, or temporal processing, is known to decline with aging and hearing loss and negatively impact auditory perception in general and speech recognition specifically. However, neurophysiological literature also provides evidence of exaggerated or enhanced encoding of specifically temporal envelopes in aging and hearing loss, which may arise from changes in inhibitory neurotransmission and neuronal hyperactivity. This review paper describes the physiological changes to the neural encoding of temporal envelopes that have been shown to occur with age and hearing loss and discusses the role of disinhibition and neural hyperactivity in contributing to these changes. Studies in both humans and animal models suggest that aging and hearing loss are associated with stronger neural representations of both periodic amplitude modulation envelopes and of naturalistic speech envelopes, but primarily for low-frequency modulations (<80 Hz). Although the frequency dependence of these results is generally taken as evidence of amplified envelope encoding at the cortex and impoverished encoding at the midbrain and brainstem, there is additional evidence to suggest that exaggerated envelope encoding may also occur subcortically, though only for envelopes with low modulation rates. A better understanding of how temporal envelope encoding is altered in aging and hearing loss, and the contexts in which neural responses are exaggerated/diminished, may aid in the development of interventions, assistive devices, and treatment strategies that work to ameliorate age- and hearing-loss-related auditory perceptual deficits.
Collapse
Affiliation(s)
- Carolyn M McClaskey
- Department of Otolaryngology - Head and Neck Surgery, Medical University of South Carolina, 135 Rutledge Ave, MSC 550, Charleston, SC 29425, United States.
| |
Collapse
|
6
|
Parameshwarappa V, Siponen M, Watabe I, Karkaba A, Galazyuk A, Noreña A. Noise-Induced Hearing Loss Alters Potassium-Chloride CoTransporter KCC2 and GABA Inhibition in the auditory centers. RESEARCH SQUARE 2023:rs.3.rs-3389804. [PMID: 37886592 PMCID: PMC10602088 DOI: 10.21203/rs.3.rs-3389804/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Homeostatic plasticity, the ability of neurons to maintain their averaged activity constant around a set point value, is thought to account for the central hyperactivity after hearing loss. Here, we investigated the putative role of GABAergic neurotransmission in this mechanism after a noise-induced hearing loss larger than 50 dB in high frequencies in guinea pigs. The effect of GABAergic inhibition is linked to the normal functioning of K+-Cl- co-transporter isoform 2 (KCC2) which maintains a low intracellular concentration of chloride. The expression of membrane KCC2 were investigated before after noise trauma in the ventral and dorsal cochlear nucleus (VCN and DCN, respectively) and in the inferior colliculus (IC). Moreover, the effect of gabazine (GBZ), a GABA antagonist, was also studied on the neural activity in IC. We show that KCC2 is downregulated in VCN, DCN and IC 3 days after noise trauma, and in DCN and IC 30 days after the trauma. As expected, GBZ application in the IC of control animals resulted in an increase of spontaneous and stimulus-evoked activity. In the noise exposed animals, on the other hand, GBZ application decreased the stimulus-evoked activity in IC neurons. The functional implications of these central changes are discussed.
Collapse
Affiliation(s)
| | - Marina Siponen
- Centre National de la Recherche Scientifique, Aix- Marseille University
| | - Isabelle Watabe
- Centre National de la Recherche Scientifique, Aix- Marseille University
| | - Alaa Karkaba
- Centre National de la Recherche Scientifique, Aix- Marseille University
| | | | - Arnaud Noreña
- Centre National de la Recherche Scientifique, Aix- Marseille University
| |
Collapse
|
7
|
Pisani A, Paciello F, Del Vecchio V, Malesci R, De Corso E, Cantone E, Fetoni AR. The Role of BDNF as a Biomarker in Cognitive and Sensory Neurodegeneration. J Pers Med 2023; 13:jpm13040652. [PMID: 37109038 PMCID: PMC10140880 DOI: 10.3390/jpm13040652] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/04/2023] [Accepted: 04/08/2023] [Indexed: 04/29/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) has a crucial function in the central nervous system and in sensory structures including olfactory and auditory systems. Many studies have highlighted the protective effects of BDNF in the brain, showing how it can promote neuronal growth and survival and modulate synaptic plasticity. On the other hand, conflicting data about BDNF expression and functions in the cochlear and in olfactory structures have been reported. Several clinical and experimental research studies showed alterations in BDNF levels in neurodegenerative diseases affecting the central and peripheral nervous system, suggesting that BDNF can be a promising biomarker in most neurodegenerative conditions, including Alzheimer's disease, shearing loss, or olfactory impairment. Here, we summarize current research concerning BDNF functions in brain and in sensory domains (olfaction and hearing), focusing on the effects of the BDNF/TrkB signalling pathway activation in both physiological and pathological conditions. Finally, we review significant studies highlighting the possibility to target BDNF as a biomarker in early diagnosis of sensory and cognitive neurodegeneration, opening new opportunities to develop effective therapeutic strategies aimed to counteract neurodegeneration.
Collapse
Affiliation(s)
- Anna Pisani
- Department of Otolaryngology Head and Neck Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Fabiola Paciello
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Valeria Del Vecchio
- Department of Neuroscience, Reproductive Sciences and Dentistry-Audiology Section, University of Naples Federico II, 80131 Naples, Italy
| | - Rita Malesci
- Department of Neuroscience, Reproductive Sciences and Dentistry-Audiology Section, University of Naples Federico II, 80131 Naples, Italy
| | - Eugenio De Corso
- Department of Otolaryngology Head and Neck Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Elena Cantone
- Department of Neuroscience, Reproductive Sciences and Dentistry-ENT Section, University of Naples Federico II, 80131 Naples, Italy
| | - Anna Rita Fetoni
- Department of Neuroscience, Reproductive Sciences and Dentistry-Audiology Section, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
8
|
Xue B, Meng X, Kao JPY, Kanold PO. Age-related changes in excitatory and inhibitory intra-cortical circuits in auditory cortex of C57Bl/6 mice. Hear Res 2023; 429:108685. [PMID: 36701895 PMCID: PMC9928889 DOI: 10.1016/j.heares.2022.108685] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 12/16/2022] [Accepted: 12/26/2022] [Indexed: 12/28/2022]
Abstract
A common impairment in aging is age-related hearing loss (presbycusis), which manifests as impaired spectrotemporal processing. Aging is accompanied by alteration in normal inhibitory (GABA) neurotransmission, and changes in excitatory (NMDA and AMPA) synapses in the auditory cortex (ACtx). However, the circuits affected by these synaptic changes remain unknown. Mice of the C57Bl/6J strain show premature age-related hearing loss and changes in functional responses in ACtx. We thus investigated how auditory cortical microcircuits change with age by comparing young (∼ 6 weeks) and aged (>1 year old) C57Bl/6J mice. We performed laser scanning photostimulation (LSPS) combined with whole-cell patch clamp recordings from Layer (L) 2/3 cells in primary auditory cortex (A1) of young adult and aged C57Bl/6J mice. We found that L2/3 cells in aged C57Bl/6J mice display functional hypoconnectivity of both excitatory and inhibitory circuits. Compared to cells from young C57Bl/6 mice, cells from aged C57Bl/6J mice have fewer excitatory connections with weaker connection strength. Whereas young adult and aged C57Bl/6J mice have similar amounts of inhibitory connections, the strength of local inhibition is weaker in the aged group. We confirmed these results by recording miniature excitatory (mEPSCs) and inhibitory synaptic currents (mIPSCs). Our results suggest a specific reduction in excitatory and inhibitory intralaminar cortical circuits in aged C57Bl/6J mice compared with young adult animals. We speculate that these unbalanced changes in cortical circuits contribute to the functional manifestations of age-related hearing loss.
Collapse
Affiliation(s)
- Binghan Xue
- Department of Biology, University of Maryland, College Park, MD 20742, United States
| | - Xiangying Meng
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, United States; Department of Biology, University of Maryland, College Park, MD 20742, United States
| | - Joseph P Y Kao
- Center for Biomedical Engineering and Technology, and Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Patrick O Kanold
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, United States; Department of Biology, University of Maryland, College Park, MD 20742, United States.
| |
Collapse
|
9
|
Can GABAkines Quiet the Noise? The GABAA Receptor Neurobiology and Pharmacology of Tinnitus. Biochem Pharmacol 2022; 201:115067. [DOI: 10.1016/j.bcp.2022.115067] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 11/20/2022]
|
10
|
Knipper M, Singer W, Schwabe K, Hagberg GE, Li Hegner Y, Rüttiger L, Braun C, Land R. Disturbed Balance of Inhibitory Signaling Links Hearing Loss and Cognition. Front Neural Circuits 2022; 15:785603. [PMID: 35069123 PMCID: PMC8770933 DOI: 10.3389/fncir.2021.785603] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/08/2021] [Indexed: 12/19/2022] Open
Abstract
Neuronal hyperexcitability in the central auditory pathway linked to reduced inhibitory activity is associated with numerous forms of hearing loss, including noise damage, age-dependent hearing loss, and deafness, as well as tinnitus or auditory processing deficits in autism spectrum disorder (ASD). In most cases, the reduced central inhibitory activity and the accompanying hyperexcitability are interpreted as an active compensatory response to the absence of synaptic activity, linked to increased central neural gain control (increased output activity relative to reduced input). We here suggest that hyperexcitability also could be related to an immaturity or impairment of tonic inhibitory strength that typically develops in an activity-dependent process in the ascending auditory pathway with auditory experience. In these cases, high-SR auditory nerve fibers, which are critical for the shortest latencies and lowest sound thresholds, may have either not matured (possibly in congenital deafness or autism) or are dysfunctional (possibly after sudden, stressful auditory trauma or age-dependent hearing loss linked with cognitive decline). Fast auditory processing deficits can occur despite maintained basal hearing. In that case, tonic inhibitory strength is reduced in ascending auditory nuclei, and fast inhibitory parvalbumin positive interneuron (PV-IN) dendrites are diminished in auditory and frontal brain regions. This leads to deficits in central neural gain control linked to hippocampal LTP/LTD deficiencies, cognitive deficits, and unbalanced extra-hypothalamic stress control. Under these conditions, a diminished inhibitory strength may weaken local neuronal coupling to homeostatic vascular responses required for the metabolic support of auditory adjustment processes. We emphasize the need to distinguish these two states of excitatory/inhibitory imbalance in hearing disorders: (i) Under conditions of preserved fast auditory processing and sustained tonic inhibitory strength, an excitatory/inhibitory imbalance following auditory deprivation can maintain precise hearing through a memory linked, transient disinhibition that leads to enhanced spiking fidelity (central neural gain⇑) (ii) Under conditions of critically diminished fast auditory processing and reduced tonic inhibitory strength, hyperexcitability can be part of an increased synchronization over a broader frequency range, linked to reduced spiking reliability (central neural gain⇓). This latter stage mutually reinforces diminished metabolic support for auditory adjustment processes, increasing the risks for canonical dementia syndromes.
Collapse
Affiliation(s)
- Marlies Knipper
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
- *Correspondence: Marlies Knipper,
| | - Wibke Singer
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Kerstin Schwabe
- Experimental Neurosurgery, Department of Neurosurgery, Hannover Medical School, Hanover, Germany
| | - Gisela E. Hagberg
- Department of Biomedical Magnetic Resonance, University Hospital Tübingen (UKT), Tübingen, Germany
- High-Field Magnetic Resonance, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Yiwen Li Hegner
- MEG Center, University of Tübingen, Tübingen, Germany
- Center of Neurology, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Lukas Rüttiger
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Christoph Braun
- MEG Center, University of Tübingen, Tübingen, Germany
- Center of Neurology, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Rüdiger Land
- Department of Experimental Otology, Institute for Audioneurotechnology, Hannover Medical School, Hanover, Germany
| |
Collapse
|
11
|
Shilling-Scrivo K, Mittelstadt J, Kanold PO. Altered Response Dynamics and Increased Population Correlation to Tonal Stimuli Embedded in Noise in Aging Auditory Cortex. J Neurosci 2021; 41:9650-9668. [PMID: 34611028 PMCID: PMC8612470 DOI: 10.1523/jneurosci.0839-21.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 09/25/2021] [Accepted: 09/29/2021] [Indexed: 11/21/2022] Open
Abstract
Age-related hearing loss (presbycusis) is a chronic health condition that affects one-third of the world population. One hallmark of presbycusis is a difficulty hearing in noisy environments. Presbycusis can be separated into two components: alterations of peripheral mechanotransduction of sound in the cochlea and central alterations of auditory processing areas of the brain. Although the effects of the aging cochlea in hearing loss have been well studied, the role of the aging brain in hearing loss is less well understood. Therefore, to examine how age-related central processing changes affect hearing in noisy environments, we used a mouse model (Thy1-GCaMP6s X CBA) that has excellent peripheral hearing in old age. We used in vivo two-photon Ca2+ imaging to measure the responses of neuronal populations in auditory cortex (ACtx) of adult (2-6 months, nine male, six female, 4180 neurons) and aging mice (15-17 months, six male, three female, 1055 neurons) while listening to tones in noisy backgrounds. We found that ACtx neurons in aging mice showed larger responses to tones and have less suppressed responses consistent with reduced inhibition. Aging neurons also showed less sensitivity to temporal changes. Population analysis showed that neurons in aging mice showed higher pairwise activity correlations and showed a reduced diversity in responses to sound stimuli. Using neural decoding techniques, we show a loss of information in neuronal populations in the aging brain. Thus, aging not only affects the responses of single neurons but also affects how these neurons jointly represent stimuli.SIGNIFICANCE STATEMENT Aging results in hearing deficits particularly under challenging listening conditions. We show that auditory cortex contains distinct subpopulations of excitatory neurons that preferentially encode different stimulus features and that aging selectively reduces certain subpopulations. We also show that aging increases correlated activity between neurons and thereby reduces the response diversity in auditory cortex. The loss of population response diversity leads to a decrease of stimulus information and deficits in sound encoding, especially in noisy backgrounds. Future work determining the identities of circuits affected by aging could provide new targets for therapeutic strategies.
Collapse
Affiliation(s)
- Kelson Shilling-Scrivo
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland 21230
| | - Jonah Mittelstadt
- Department of Biology, University of Maryland, College Park, Maryland 20742
| | - Patrick O Kanold
- Department of Biology, University of Maryland, College Park, Maryland 20742
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 20215
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD 21205
| |
Collapse
|
12
|
Paciello F, Rinaudo M, Longo V, Cocco S, Conforto G, Pisani A, Podda MV, Fetoni AR, Paludetti G, Grassi C. Auditory sensory deprivation induced by noise exposure exacerbates cognitive decline in a mouse model of Alzheimer's disease. eLife 2021; 10:70908. [PMID: 34699347 PMCID: PMC8547960 DOI: 10.7554/elife.70908] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 10/15/2021] [Indexed: 12/18/2022] Open
Abstract
Although association between hearing impairment and dementia has been widely documented by epidemiological studies, the role of auditory sensory deprivation in cognitive decline remains to be fully understood. To address this issue we investigated the impact of hearing loss on the onset and time-course of cognitive decline in an animal model of Alzheimer's disease (AD), that is the 3×Tg-AD mice and the underlying mechanisms. We found that hearing loss induced by noise exposure in the 3×Tg-AD mice before the phenotype is manifested caused persistent synaptic and morphological alterations in the auditory cortex. This was associated with earlier hippocampal dysfunction, increased tau phosphorylation, neuroinflammation, and redox imbalance, along with anticipated memory deficits compared to the expected time-course of the neurodegenerative phenotype. Our data suggest that a mouse model of AD is more vulnerable to central damage induced by hearing loss and shows reduced ability to counteract noise-induced detrimental effects, which accelerates the neurodegenerative disease onset.
Collapse
Affiliation(s)
- Fabiola Paciello
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marco Rinaudo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Longo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Sara Cocco
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giulia Conforto
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Anna Pisani
- Department of Otolaryngology Head and Neck Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maria Vittoria Podda
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Anna Rita Fetoni
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Otolaryngology Head and Neck Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gaetano Paludetti
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Otolaryngology Head and Neck Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Claudio Grassi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
13
|
Salvi R, Radziwon K, Manohar S, Auerbach B, Ding D, Liu X, Lau C, Chen YC, Chen GD. Review: Neural Mechanisms of Tinnitus and Hyperacusis in Acute Drug-Induced Ototoxicity. Am J Audiol 2021; 30:901-915. [PMID: 33465315 DOI: 10.1044/2020_aja-20-00023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Purpose Tinnitus and hyperacusis are debilitating conditions often associated with age-, noise-, and drug-induced hearing loss. Because of their subjective nature, the neural mechanisms that give rise to tinnitus and hyperacusis are poorly understood. Over the past few decades, considerable progress has been made in deciphering the biological bases for these disorders using animal models. Method Important advances in understanding the biological bases of tinnitus and hyperacusis have come from studies in which tinnitus and hyperacusis are consistently induced with a high dose of salicylate, the active ingredient in aspirin. Results Salicylate induced a transient hearing loss characterized by a reduction in otoacoustic emissions, a moderate cochlear threshold shift, and a large reduction in the neural output of the cochlea. As the weak cochlear neural signals were relayed up the auditory pathway, they were progressively amplified so that the suprathreshold neural responses in the auditory cortex were much larger than normal. Excessive central gain (neural amplification), presumably resulting from diminished inhibition, is believed to contribute to hyperacusis and tinnitus. Salicylate also increased corticosterone stress hormone levels. Functional imaging studies indicated that salicylate increased spontaneous activity and enhanced functional connectivity between structures in the central auditory pathway and regions of the brain associated with arousal (reticular formation), emotion (amygdala), memory/spatial navigation (hippocampus), motor planning (cerebellum), and motor control (caudate/putamen). Conclusion These results suggest that tinnitus and hyperacusis arise from aberrant neural signaling in a complex neural network that includes both auditory and nonauditory structures.
Collapse
Affiliation(s)
- Richard Salvi
- Center for Hearing & Deafness, Department of Communicative Disorders and Sciences, The State University of New York at Buffalo
| | - Kelly Radziwon
- Center for Hearing & Deafness, Department of Communicative Disorders and Sciences, The State University of New York at Buffalo
| | - Senthilvelan Manohar
- Center for Hearing & Deafness, Department of Communicative Disorders and Sciences, The State University of New York at Buffalo
| | - Ben Auerbach
- Center for Hearing & Deafness, Department of Communicative Disorders and Sciences, The State University of New York at Buffalo
| | - Dalian Ding
- Center for Hearing & Deafness, Department of Communicative Disorders and Sciences, The State University of New York at Buffalo
| | - Xiaopeng Liu
- Center for Hearing & Deafness, Department of Communicative Disorders and Sciences, The State University of New York at Buffalo
| | - Condon Lau
- Department of Physics, City University of Hong Kong
| | - Yu-Chen Chen
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, China
| | - Guang-Di Chen
- Center for Hearing & Deafness, Department of Communicative Disorders and Sciences, The State University of New York at Buffalo
| |
Collapse
|
14
|
Cerrah Gunes M, Gunes MS, Vural A, Aybuga F, Bayram A, Bayram KK, Sahin MI, Dogan ME, Ozdemir SY, Ozkul Y. Change in gene expression levels of GABA, glutamate and neurosteroid pathways due to acoustic trauma in the cochlea. J Neurogenet 2021; 35:45-57. [PMID: 33825593 DOI: 10.1080/01677063.2021.1904922] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The characteristic feature of noise-induced hearing loss (NIHL) is the loss or malfunction of the outer hair cells (OHC) and the inner hair cells (IHC) of the cochlea. 90-95% of the spiral ganglion neurons, forming the cell bodies of cochlear nerve, synapse with the IHCs. Glutamate is the most potent excitatory neurotransmitter for IHC-auditory nerve synapses. Excessive release of glutamate in response to acoustic trauma (AT), may cause excitotoxicity by causing damage to the spiral ganglion neurons (SGN) or loss of the spiral ganglion dendrites, post-synaptic to the IHCs. Another neurotransmitter, GABA, plays an important role in the processing of acoustic stimuli and central regulation after peripheral injury, so it is potentially related to the regulation of hearing function and sensitivity after noise. The aim of this study is to evaluate the effect of AT on the expressions of glutamate excitotoxicity, GABA inhibition and neurosteroid synthesis genes.We exposed 24 BALB/c mice to AT. Controls were sacrificed without exposure to noise, Post-AT(1) and Post-AT(15) were sacrificed on the 1st and 15th day, respectively, after noise exposure. The expressions of various genes playing roles in glutamate, GABA and neurosteroid pathways were compared between groups by real-time PCR.Expressions of Cyp11a1, Gls, Gabra1, Grin2b, Sult1a1, Gad1, and Slc1a2 genes in Post-AT(15) mice were significantly decreased in comparison to control and Post-AT(1) mice. No significant differences in the expression of Slc6a1 and Slc17a8 genes was detected.These findings support the possible role of balance between glutamate excitotoxicity and GABA inhibition is disturbed during the post AT days and also the synthesis of some neurosteroids such as pregnenolone sulfate may be important in this balance.
Collapse
Affiliation(s)
- Meltem Cerrah Gunes
- Department of Medical Genetics, School of Medicine, Erciyes University, Kayseri, Turkey
| | - Murat Salih Gunes
- Department of Otolaryngology, Izmit Seka State Hospital, Kocaeli, Turkey
| | - Alperen Vural
- Department of Otolaryngology, School of Medicine, Erciyes University, Kayseri, Turkey
| | | | - Arslan Bayram
- Etlik Zübeyde Hanım Women's Diseases Education and Research Hospital, Health Sciences University, T.R. Ministry of Health, Ankara, Turkey
| | - Keziban Korkmaz Bayram
- Department of Medical Genetics, School of Medicine, Yıldirim Beyazit University, Ankara, Turkey
| | - Mehmet Ilhan Sahin
- Department of Otolaryngology, School of Medicine, Erciyes University, Kayseri, Turkey
| | - Muhammet Ensar Dogan
- Department of Medical Genetics, School of Medicine, Erciyes University, Kayseri, Turkey
| | - Sevda Yesim Ozdemir
- Department of Medical Genetics, School of Medicine, Uskudar University, Istanbul, Turkey
| | - Yusuf Ozkul
- Department of Medical Genetics, School of Medicine, Erciyes University, Kayseri, Turkey.,Center of Genome and Stem Cell, Kayseri, Turkey
| |
Collapse
|
15
|
Luan Y, Salvi R, Liu L, Lu C, Jiao Y, Tang T, Liu H, Teng GJ. High-frequency Noise-induced Hearing Loss Disrupts Functional Connectivity in Non-auditory Areas with Cognitive Disturbances. Neurosci Bull 2021; 37:720-724. [PMID: 33772721 DOI: 10.1007/s12264-021-00663-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 12/02/2020] [Indexed: 10/21/2022] Open
Affiliation(s)
- Ying Luan
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Richard Salvi
- Center for Hearing and Deafness, University at Buffalo, State University of New York, Buffalo, 14214, USA
| | - Lijie Liu
- Department of Physiology, Medical College of Southeast University, Nanjing, 210009, China
| | - Chunqiang Lu
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Yun Jiao
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Tianyu Tang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Haiqing Liu
- Department of Physiology, Medical College of Southeast University, Nanjing, 210009, China
| | - Gao-Jun Teng
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China.
| |
Collapse
|
16
|
Henton A, Tzounopoulos T. What's the buzz? The neuroscience and the treatment of tinnitus. Physiol Rev 2021; 101:1609-1632. [PMID: 33769102 DOI: 10.1152/physrev.00029.2020] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Tinnitus is a pervasive public health issue that affects ∼15% of the United States population. Similar estimates have also been shown on a global scale, with similar prevalence found in Europe, Asia, and Africa. The severity of tinnitus is heterogeneous, ranging from mildly bothersome to extremely disruptive. In the United States, ∼10-20% of individuals who experience tinnitus report symptoms that severely reduce their quality of life. Due to the huge personal and societal burden, in the last 20 yr a concerted effort on basic and clinical research has significantly advanced our understanding and treatment of this disorder. Yet, neither full understanding, nor cure exists. We know that tinnitus is the persistent involuntary phantom percept of internally generated nonverbal indistinct noises and tones, which in most cases is initiated by acquired hearing loss and maintained only when this loss is coupled with distinct neuronal changes in auditory and extra-auditory brain networks. Yet, the exact mechanisms and patterns of neural activity that are necessary and sufficient for the perceptual generation and maintenance of tinnitus remain incompletely understood. Combinations of animal model and human research will be essential in filling these gaps. Nevertheless, the existing progress in investigating the neurophysiological mechanisms has improved current treatment and highlighted novel targets for drug development and clinical trials. The aim of this review is to thoroughly discuss the current state of human and animal tinnitus research, outline current challenges, and highlight new and exciting research opportunities.
Collapse
Affiliation(s)
- A Henton
- Pittsburgh Hearing Research Center and Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - T Tzounopoulos
- Pittsburgh Hearing Research Center and Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
17
|
The Neural Bases of Tinnitus: Lessons from Deafness and Cochlear Implants. J Neurosci 2021; 40:7190-7202. [PMID: 32938634 DOI: 10.1523/jneurosci.1314-19.2020] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 08/05/2020] [Accepted: 08/08/2020] [Indexed: 02/06/2023] Open
Abstract
Subjective tinnitus is the conscious perception of sound in the absence of any acoustic source. The literature suggests various tinnitus mechanisms, most of which invoke changes in spontaneous firing rates of central auditory neurons resulting from modification of neural gain. Here, we present an alternative model based on evidence that tinnitus is: (1) rare in people who are congenitally deaf, (2) common in people with acquired deafness, and (3) potentially suppressed by active cochlear implants used for hearing restoration. We propose that tinnitus can only develop after fast auditory fiber activity has stimulated the synapse formation between fast-spiking parvalbumin positive (PV+) interneurons and projecting neurons in the ascending auditory path and coactivated frontostriatal networks after hearing onset. Thereafter, fast auditory fiber activity promotes feedforward and feedback inhibition mediated by PV+ interneuron activity in auditory-specific circuits. This inhibitory network enables enhanced stimulus resolution, attention-driven contrast improvement, and augmentation of auditory responses in central auditory pathways (neural gain) after damage of slow auditory fibers. When fast auditory fiber activity is lost, tonic PV+ interneuron activity is diminished, resulting in the prolonged response latencies, sudden hyperexcitability, enhanced cortical synchrony, elevated spontaneous γ oscillations, and impaired attention/stress-control that have been described in previous tinnitus models. Moreover, because fast processing is gained through sensory experience, tinnitus would not exist in congenital deafness. Electrical cochlear stimulation may have the potential to reestablish tonic inhibitory networks and thus suppress tinnitus. The proposed framework unites many ideas of tinnitus pathophysiology and may catalyze cooperative efforts to develop tinnitus therapies.
Collapse
|
18
|
Zinsmaier AK, Wang W, Zhang L, Hossainy NN, Bao S. Resistance to noise-induced gap detection impairment in FVB mice is correlated with reduced neuroinflammatory response and parvalbumin-positive neuron loss. Sci Rep 2020; 10:20445. [PMID: 33235216 PMCID: PMC7686384 DOI: 10.1038/s41598-020-75714-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 10/12/2020] [Indexed: 01/10/2023] Open
Abstract
Exposure to loud noises results in neuroinflammatory responses in the central auditory pathway. Noise-induced neuroinflammation is implicated in auditory processing deficits such as impairment in gap detection. In this study, we examined whether strain differences between the FVB and C57BL/6 mice in noise-induced impairment in gap detection are correlated with strain differences in neuroinflammatory responses. We found that noise induced more robust TNF-α expression in C57BL/6 than in FVB mice. Noise-induced microglial deramification was observed in C57BL/6 mice, but not in FVB mice. Furthermore, noise exposure resulted in a reduction in parvalbumin-positive (PV+) neuron density in the C57BL/6 mice, but not in FVB mice. These results suggest that neuroinflammatory responses and loss of PV+ neurons may contribute to strain differences in noise-induced impairment in gap detection.
Collapse
Affiliation(s)
- Alexander K Zinsmaier
- Department of Physiology, University of Arizona College of Medicine, Tucson, AZ, 85724, USA
| | - Weihua Wang
- Department of Physiology, University of Arizona College of Medicine, Tucson, AZ, 85724, USA
| | - Li Zhang
- Department of Physiology, University of Arizona College of Medicine, Tucson, AZ, 85724, USA
| | - Nadia N Hossainy
- Department of Physiology, University of Arizona College of Medicine, Tucson, AZ, 85724, USA
| | - Shaowen Bao
- Department of Physiology, University of Arizona College of Medicine, Tucson, AZ, 85724, USA.
| |
Collapse
|
19
|
Encoding of a binaural speech stimulus at the brainstem level in middle-aged adults. The Journal of Laryngology & Otology 2020; 134:1044-1051. [PMID: 33153510 DOI: 10.1017/s0022215120002285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVE Binaural hearing is facilitated by neural interactions in the auditory pathway. Ageing results in impairment of localisation and listening in noisy situations without any significant hearing loss. The present study focused on comparing the binaural encoding of a speech stimulus at the subcortical level in middle-aged versus younger adults, based on speech-evoked auditory brainstem responses. METHODS Thirty participants (15 young adults and 15 middle-aged adults) with normal hearing sensitivity (less than 15 dB HL) participated in the study. The speech-evoked auditory brainstem response was recorded monaurally and binaurally with a 40-ms /da/ stimulus. Fast Fourier transform analysis was utilised. RESULTS An independent sample t-test revealed a significant difference between the two groups in fundamental frequency (F0) amplitude recorded with binaural stimulation. CONCLUSION The present study suggested that ageing results in degradation of F0 encoding, which is essential for the perception of speech in noise.
Collapse
|
20
|
Deng D, Wang W, Bao S. Diffusible Tumor Necrosis Factor-Alpha (TNF-α) Promotes Noise-Induced Parvalbumin-Positive (PV+) Neuron Loss and Auditory Processing Impairments. Front Neurosci 2020; 14:573047. [PMID: 33154715 PMCID: PMC7590827 DOI: 10.3389/fnins.2020.573047] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/15/2020] [Indexed: 12/11/2022] Open
Abstract
Neuroinflammation has been implicated in noise-induced auditory processing disorder and tinnitus. Certain non-auditory neurological disorders can also increase the levels of proinflammatory cytokines in the brain. To investigate the impact of increased brain proinflammatory cytokine levels on the central auditory pathway, we infused recombinant TNF-α into the right lateral cerebral ventricle, and examined auditory processing and cytoarchitecture of the auditory cortex. Microglial deramification was observed in the auditory cortex of mice that had received both TNF-α infusion and exposure to an 86-dB noise, but not in mice that had received either TNF-α infusion or noise exposure alone. In addition, we observed reduced cortical PV+ neuron density and impaired performances in gap detection and prepulse inhibition (PPI) only in mice that received both TNF-α infusion and the noise exposure. These results suggest that disease-related increase in brain proinflammatory cytokine release could be a risk factor for noise-induced auditory processing disorder and tinnitus.
Collapse
Affiliation(s)
- Di Deng
- Department of Physiology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Weihua Wang
- Department of Physiology, College of Medicine, University of Arizona, Tucson, AZ, United States
| | - Shaowen Bao
- Department of Physiology, College of Medicine, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
21
|
Wong E, Radziwon K, Chen GD, Liu X, Manno FA, Manno SH, Auerbach B, Wu EX, Salvi R, Lau C. Functional magnetic resonance imaging of enhanced central auditory gain and electrophysiological correlates in a behavioral model of hyperacusis. Hear Res 2020; 389:107908. [PMID: 32062293 DOI: 10.1016/j.heares.2020.107908] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/02/2019] [Accepted: 02/02/2020] [Indexed: 01/24/2023]
Abstract
Hyperacusis is a debilitating hearing condition in which normal everyday sounds are perceived as exceedingly loud, annoying, aversive or even painful. The prevalence of hyperacusis approaches 10%, making it an important, but understudied medical condition. To noninvasively identify the neural correlates of hyperacusis in an animal model, we used sound-evoked functional magnetic resonance imaging (fMRI) to locate regions of abnormal activity in the central nervous system of rats with behavioral evidence of hyperacusis induced with an ototoxic drug (sodium salicylate, 250 mg/kg, i.p.). Reaction time-intensity measures of loudness-growth revealed behavioral evidence of salicylate-induced hyperacusis at high intensities. fMRI revealed significantly enhanced sound-evoked responses in the auditory cortex (AC) to 80 dB SPL tone bursts presented at 8 and 16 kHz. Sound-evoked responses in the inferior colliculus (IC) were also enhanced, but to a lesser extent. To confirm the main results, electrophysiological recordings of spike discharges from multi-unit clusters were obtained from the central auditory pathway. Salicylate significantly enhanced tone-evoked spike-discharges from multi-unit clusters in the AC from 4 to 30 kHz at intensities ≥60 dB SPL; less enhancement occurred in the medial geniculate body (MGB), and even less in the IC. Our results demonstrate for the first time that non-invasive sound-evoked fMRI can be used to identify regions of neural hyperactivity throughout the brain in an animal model of hyperacusis.
Collapse
Affiliation(s)
- Eddie Wong
- Department of Physics, City University of Hong Kong, Hong Kong, China; Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China; Laboratory of Biomedical Imaging and Signal Processing, The University of Hong Kong, Hong Kong, China
| | - Kelly Radziwon
- Center for Hearing & Deafness, Department of Communicative Disorders and Sciences, SUNY at Buffalo, 137 Cary Hall, Buffalo, NY, 14214, USA
| | - Guang-Di Chen
- Center for Hearing & Deafness, Department of Communicative Disorders and Sciences, SUNY at Buffalo, 137 Cary Hall, Buffalo, NY, 14214, USA
| | - Xiaopeng Liu
- Center for Hearing & Deafness, Department of Communicative Disorders and Sciences, SUNY at Buffalo, 137 Cary Hall, Buffalo, NY, 14214, USA
| | - Francis Am Manno
- Department of Physics, City University of Hong Kong, Hong Kong, China; School of Biomedical Engineering, University of Sydney, Sydney, New South Wales, Australia
| | - Sinai Hc Manno
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Benjamin Auerbach
- Center for Hearing & Deafness, Department of Communicative Disorders and Sciences, SUNY at Buffalo, 137 Cary Hall, Buffalo, NY, 14214, USA
| | - Ed X Wu
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong, China; Laboratory of Biomedical Imaging and Signal Processing, The University of Hong Kong, Hong Kong, China
| | - Richard Salvi
- Center for Hearing & Deafness, Department of Communicative Disorders and Sciences, SUNY at Buffalo, 137 Cary Hall, Buffalo, NY, 14214, USA; Department of Audiology and Speech-Language Pathology, Asia University, Taichung, Taiwan, ROC.
| | - Condon Lau
- Department of Physics, City University of Hong Kong, Hong Kong, China.
| |
Collapse
|
22
|
Salvi R, Auerbach BD, Lau C, Chen YC, Manohar S, Liu X, Ding D, Chen GD. Functional Neuroanatomy of Salicylate- and Noise-Induced Tinnitus and Hyperacusis. Curr Top Behav Neurosci 2020; 51:133-160. [PMID: 32653998 DOI: 10.1007/7854_2020_156] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Tinnitus and hyperacusis are debilitating conditions often associated with aging or exposure to intense noise or ototoxic drugs. One of the most reliable methods of inducing tinnitus is with high doses of sodium salicylate, the active ingredient in aspirin. High doses of salicylate have been widely used to investigate the functional neuroanatomy of tinnitus and hyperacusis. High doses of salicylate have been used to develop novel behavioral methods to detect the presence of tinnitus and hyperacusis in animal models. Salicylate typically induces a hearing loss of approximately 20 dB which greatly reduces the neural output of the cochlea. As this weak neural signal emerging from the cochlea is sequentially relayed to the cochlear nucleus, inferior colliculus, medial geniculate, and auditory cortex, the neural response to suprathreshold sounds is progressively amplified by a factor of 2-3 by the time the signal reaches the auditory cortex, a phenomenon referred to as enhanced central gain. Sound-evoked hyperactivity also occurred in the amygdala, a region that assigns emotional significance to sensory stimuli. Resting state functional magnetic imaging of the BOLD signal revealed salicylate-induced increases in spontaneous neural activity in the inferior colliculus, medial geniculate body, and auditory cortex as well as in non-auditory areas such as the amygdala, reticular formation, cerebellum, and other sensory areas. Functional connectivity of the BOLD signal revealed increased neural coupling between several auditory areas and non-auditory areas such as the amygdala, cerebellum, reticular formation, hippocampus, and caudate/putamen; these strengthened connections likely contribute to the multifaceted dimensions of tinnitus. Taken together, these results suggest that salicylate-induced tinnitus disrupts a complex neural network involving many auditory centers as well as brain regions involved with emotion, arousal, memory, and motor planning. These extra-auditory centers embellish the basic auditory percepts that results in tinnitus and which may also contribute to hyperacusis.
Collapse
Affiliation(s)
- Richard Salvi
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, USA.
| | | | - Condon Lau
- Department of Physics, City University of Hong Kong, Hong Kong, China
| | - Yu-Chen Chen
- Department of Radiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | | | - Xiaopeng Liu
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, USA
| | - Dalian Ding
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, USA
| | - Guang-Di Chen
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
23
|
Ibrahim BA, Llano DA. Aging and Central Auditory Disinhibition: Is It a Reflection of Homeostatic Downregulation or Metabolic Vulnerability? Brain Sci 2019; 9:brainsci9120351. [PMID: 31805729 PMCID: PMC6955996 DOI: 10.3390/brainsci9120351] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 11/25/2019] [Accepted: 11/30/2019] [Indexed: 01/08/2023] Open
Abstract
Aging-related changes have been identified at virtually every level of the central auditory system. One of the most common findings across these nuclei is a loss of synaptic inhibition with aging, which has been proposed to be at the heart of several aging-related changes in auditory cognition, including diminished speech perception in complex environments and the presence of tinnitus. Some authors have speculated that downregulation of synaptic inhibition is a consequence of peripheral deafferentation and therefore is a homeostatic mechanism to restore excitatory/inhibitory balance. As such, disinhibition would represent a form of maladaptive plasticity. However, clinical data suggest that deafferentation-related disinhibition tends to occur primarily in the aged brain. Therefore, aging-related disinhibition may, in part, be related to the high metabolic demands of inhibitory neurons relative to their excitatory counterparts. These findings suggest that both deafferentation-related maladaptive plastic changes and aging-related metabolic factors combine to produce changes in central auditory function. Here, we explore the arguments that downregulation of inhibition may be due to homeostatic responses to diminished afferent input vs. metabolic vulnerability of inhibitory neurons in the aged brain. Understanding the relative importance of these mechanisms will be critical for the development of treatments for the underlying causes of aging-related central disinhibition.
Collapse
Affiliation(s)
- Baher A. Ibrahim
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA;
| | - Daniel A. Llano
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA;
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Correspondence:
| |
Collapse
|
24
|
Radziwon K, Auerbach BD, Ding D, Liu X, Chen GD, Salvi R. Noise-Induced loudness recruitment and hyperacusis: Insufficient central gain in auditory cortex and amygdala. Neuroscience 2019; 422:212-227. [PMID: 31669363 PMCID: PMC6994858 DOI: 10.1016/j.neuroscience.2019.09.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 09/05/2019] [Accepted: 09/10/2019] [Indexed: 12/12/2022]
Abstract
Noise-induced hearing loss generally induces loudness recruitment, but sometimes gives rise to hyperacusis, a debilitating condition in which moderate intensity sounds are perceived abnormally loud. In an attempt to develop an animal model of loudness hyperacusis, we exposed rats to a 16-20 kHz noise at 104 dB SPL for 12 weeks. Behavioral reaction time-intensity functions were used to assess loudness growth functions before, during and 2-months post-exposure. During the exposure, loudness recruitment (R) was present in the region of hearing loss, but subtle evidence of hyperacusis (H) started to emerge at the border of the hearing loss. Unexpectedly, robust evidence of hyperacusis appeared below and near the edge of the hearing loss 2-months post-exposure. To identify the neural correlates of hyperacusis and test the central gain model of hyperacusis, we recorded population neural responses from the cochlea, auditory cortex and lateral amygdala 2-months post-exposure. Compared to controls, the neural output of the cochlea was greatly reduced in the noise group. Consistent with central gain models, the gross neural responses from the auditory cortex and amygdala were proportionately much larger than those from the cochlea. However, despite central amplification, the population responses in the auditory cortex and amygdala were still below the level needed to fully account for hyperacusis and/or recruitment. Having developed procedures that can consistently induce hyperacusis in rats, our results set the stage for future studies that seek to identify the neurobiological events that give rise to hyperacusis and to develop new therapies to treat this debilitating condition.
Collapse
Affiliation(s)
- Kelly Radziwon
- Center for Hearing and Deafness, SUNY at Buffalo, Buffalo, NY 14214, USA
| | | | - Dalian Ding
- Center for Hearing and Deafness, SUNY at Buffalo, Buffalo, NY 14214, USA
| | - Xiaopeng Liu
- Center for Hearing and Deafness, SUNY at Buffalo, Buffalo, NY 14214, USA
| | - Guang-Di Chen
- Center for Hearing and Deafness, SUNY at Buffalo, Buffalo, NY 14214, USA.
| | - Richard Salvi
- Center for Hearing and Deafness, SUNY at Buffalo, Buffalo, NY 14214, USA
| |
Collapse
|
25
|
Tinnitus Correlates with Downregulation of Cortical Glutamate Decarboxylase 65 Expression But Not Auditory Cortical Map Reorganization. J Neurosci 2019; 39:9989-10001. [PMID: 31704784 DOI: 10.1523/jneurosci.1117-19.2019] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 10/23/2019] [Accepted: 11/04/2019] [Indexed: 12/13/2022] Open
Abstract
Hearing loss is the biggest risk factor for tinnitus, and hearing-loss-related pathological changes in the auditory pathway have been hypothesized as the mechanism underlying tinnitus. However, due to the comorbidity of tinnitus and hearing loss, it has been difficult to differentiate between neural correlates of tinnitus and consequences of hearing loss. In this study, we dissociated tinnitus and hearing loss in FVB mice, which exhibit robust resistance to tinnitus following monaural noise-induced hearing loss. Furthermore, knock-down of glutamate decarboxylase 65 (GAD65) expression in auditory cortex (AI) by RNA interference gave rise to tinnitus in normal-hearing FVB mice. We found that tinnitus was significantly correlated with downregulation of GAD65 in the AI. By contrast, cortical map distortions, which have been hypothesized as a mechanism underlying tinnitus, were correlated with hearing loss but not tinnitus. Our findings suggest new strategies for the rehabilitation of tinnitus and other phantom sensation, such as phantom pain.SIGNIFICANCE STATEMENT Hearing loss is the biggest risk factor for tinnitus in humans. Most animal models of tinnitus also exhibit comorbid hearing loss, making it difficult to dissociate the mechanisms underlying tinnitus from mere consequences of hearing loss. Here we show that, although both C57BL/6 and FVB mice exhibited similar noise-induced hearing threshold increase, only C57BL/6, but not FVB, mice developed tinnitus following noise exposure. Although both strains showed frequency map reorganization following noise-induced hearing loss, only C57BL/6 mice had reduced glutamate decarboxylase 65 (GAD65) expression in the auditory cortex (AI). Knocking down GAD65 expression in the AI resulted in tinnitus in normal-hearing FVB mice. Our results suggest that reduced inhibitory neuronal function, but not sensory map reorganization, underlies noise-induced tinnitus.
Collapse
|
26
|
Escabi CD, Frye MD, Trevino M, Lobarinas E. The rat animal model for noise-induced hearing loss. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2019; 146:3692. [PMID: 31795685 PMCID: PMC7480078 DOI: 10.1121/1.5132553] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Rats make excellent models for the study of medical, biological, genetic, and behavioral phenomena given their adaptability, robustness, survivability, and intelligence. The rat's general anatomy and physiology of the auditory system is similar to that observed in humans, and this has led to their use for investigating the effect of noise overexposure on the mammalian auditory system. The current paper provides a review of the rat model for studying noise-induced hearing loss and highlights advancements that have been made using the rat, particularly as these pertain to noise dose and the hazardous effects of different experimental noise types. In addition to the traditional loss of auditory function following acoustic trauma, recent findings have indicated the rat as a useful model in observing alterations in neuronal processing within the central nervous system following noise injury. Furthermore, the rat provides a second animal model when investigating noise-induced cochlear synaptopathy, as studies examining this in the rat model resemble the general patterns observed in mice. Together, these findings demonstrate the relevance of this animal model for furthering the authors' understanding of the effects of noise on structural, anatomical, physiological, and perceptual aspects of hearing.
Collapse
Affiliation(s)
- Celia D Escabi
- Callier Center for Communication Disorders, School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, Texas 75080, USA
| | - Mitchell D Frye
- Callier Center for Communication Disorders, School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, Texas 75080, USA
| | - Monica Trevino
- Callier Center for Communication Disorders, School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, Texas 75080, USA
| | - Edward Lobarinas
- Callier Center for Communication Disorders, School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, Texas 75080, USA
| |
Collapse
|
27
|
Sheppard A, Stocking C, Ralli M, Salvi R. A review of auditory gain, low-level noise and sound therapy for tinnitus and hyperacusis. Int J Audiol 2019; 59:5-15. [DOI: 10.1080/14992027.2019.1660812] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Adam Sheppard
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, USA
- Department of Communicative Disorders and Sciences, University at Buffalo, Buffalo, NY, USA
| | - Christina Stocking
- Department of Communicative Disorders and Sciences, University at Buffalo, Buffalo, NY, USA
| | - Massimo Ralli
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, USA
- Department of Communicative Disorders and Sciences, University at Buffalo, Buffalo, NY, USA
- Department of Oral and Maxillofacial Sciences, Sapienza University of Rome, Rome, Italy
| | - Richard Salvi
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, USA
- Department of Communicative Disorders and Sciences, University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
28
|
Illing RB, Buschky H, Tadic A. Mitotic activity, modulation of DNA processing, and purinergic signalling in the adult rat auditory brainstem following sensory deafferentation. Eur J Neurosci 2019; 50:3985-4003. [PMID: 31325398 DOI: 10.1111/ejn.14521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 06/13/2019] [Accepted: 07/15/2019] [Indexed: 11/30/2022]
Abstract
A complex scenario of cellular network reorganization is caused by unilateral sensory deafferentation (USD) in the adult rat central auditory system. We asked whether this plasticity response involves mitosis. Immunohistochemistry was applied to brainstem sections for the detection and localization of mitotic markers Ki67 and PCNA, the growth-associated protein Gap43 and purine receptor P2X4. Fluorescent double staining was done for Ki67:PCNA and for both of them with HuC/HuD (neurons), S100 (astrocytes), Iba1 (microglia) and P2X4. Inquiring 1-7 days after USD, we found Ki67 expression to be changed in cellular profiles of cochlear nucleus (CN) with a significant increase in number by 1-3 days, followed by reset to control level within 1 week. USD-induced mitosis exclusively occurred in microglia and was absent elsewhere in the auditory brainstem. PCNA staining of small cellular profiles increased similarly but remained elevated. PCNA staining intensity also changed in CN, superior olive and inferior colliculus in neuronal nuclei, suggesting shifts in DNA processing. No apoptotic cell death was detected in any region of the adult auditory brainstem after USD. A comparison of anterograde and retrograde effects of nerve damage revealed proliferating microglia expressing P2X4 receptors in CN upon USD, but not in the facial nucleus after facial nerve transection. In conclusion, the deafferentation model studied here permits insight into the capacity of the adult mammalian brain to invoke mitosis among glia cells, adjustment of gene processing in neurons and purinergic signalling between them, jointly accounting for a multilayered neuro- and glioplastic response.
Collapse
Affiliation(s)
- Robert-Benjamin Illing
- Neurobiological Research Laboratory, Section for Clinical-Experimental Otology, Department of Otorhinolaryngology, University Medical Center, University of Freiburg, Freiburg, Germany
| | - Helena Buschky
- Neurobiological Research Laboratory, Section for Clinical-Experimental Otology, Department of Otorhinolaryngology, University Medical Center, University of Freiburg, Freiburg, Germany
| | - Annamaria Tadic
- Neurobiological Research Laboratory, Section for Clinical-Experimental Otology, Department of Otorhinolaryngology, University Medical Center, University of Freiburg, Freiburg, Germany
| |
Collapse
|
29
|
Wang W, Zhang LS, Zinsmaier AK, Patterson G, Leptich EJ, Shoemaker SL, Yatskievych TA, Gibboni R, Pace E, Luo H, Zhang J, Yang S, Bao S. Neuroinflammation mediates noise-induced synaptic imbalance and tinnitus in rodent models. PLoS Biol 2019; 17:e3000307. [PMID: 31211773 PMCID: PMC6581239 DOI: 10.1371/journal.pbio.3000307] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 05/16/2019] [Indexed: 12/20/2022] Open
Abstract
Hearing loss is a major risk factor for tinnitus, hyperacusis, and central auditory processing disorder. Although recent studies indicate that hearing loss causes neuroinflammation in the auditory pathway, the mechanisms underlying hearing loss–related pathologies are still poorly understood. We examined neuroinflammation in the auditory cortex following noise-induced hearing loss (NIHL) and its role in tinnitus in rodent models. Our results indicate that NIHL is associated with elevated expression of proinflammatory cytokines and microglial activation—two defining features of neuroinflammatory responses—in the primary auditory cortex (AI). Genetic knockout of tumor necrosis factor alpha (TNF-α) or pharmacologically blocking TNF-α expression prevented neuroinflammation and ameliorated the behavioral phenotype associated with tinnitus in mice with NIHL. Conversely, infusion of TNF-α into AI resulted in behavioral signs of tinnitus in both wild-type and TNF-α knockout mice with normal hearing. Pharmacological depletion of microglia also prevented tinnitus in mice with NIHL. At the synaptic level, the frequency of miniature excitatory synaptic currents (mEPSCs) increased and that of miniature inhibitory synaptic currents (mIPSCs) decreased in AI pyramidal neurons in animals with NIHL. This excitatory-to-inhibitory synaptic imbalance was completely prevented by pharmacological blockade of TNF-α expression. These results implicate neuroinflammation as a therapeutic target for treating tinnitus and other hearing loss–related disorders. Prolonged exposure to loud noises causes neuronal hyperexcitability and increases the risk of tinnitus. This study reveals that this type of tinnitus is mediated by noise-induced neuroinflammation; blockade of neuroinflammatory responses prevents noise-induced neuronal excitation/inhibition imbalance and tinnitus.
Collapse
Affiliation(s)
- Weihua Wang
- Department of Physiology, University of Arizona, Tucson, Arizona, United States of America
| | - Li. S. Zhang
- Department of Physiology, University of Arizona, Tucson, Arizona, United States of America
- Helen Wills Neuroscience Institute, University of California, Berkeley, California, United States of America
| | - Alexander K. Zinsmaier
- Department of Physiology, University of Arizona, Tucson, Arizona, United States of America
| | - Genevieve Patterson
- Department of Physiology, University of Arizona, Tucson, Arizona, United States of America
| | - Emily Jean Leptich
- Department of Physiology, University of Arizona, Tucson, Arizona, United States of America
| | - Savannah L. Shoemaker
- Department of Physiology, University of Arizona, Tucson, Arizona, United States of America
| | - Tatiana A. Yatskievych
- Department of Physiology, University of Arizona, Tucson, Arizona, United States of America
| | - Robert Gibboni
- Helen Wills Neuroscience Institute, University of California, Berkeley, California, United States of America
| | - Edward Pace
- Department of Otolaryngology, Wayne State University, Detroit, Michigan, United States of America
| | - Hao Luo
- Department of Otolaryngology, Wayne State University, Detroit, Michigan, United States of America
| | - Jinsheng Zhang
- Department of Otolaryngology, Wayne State University, Detroit, Michigan, United States of America
- Department of Communication Sciences and Disorders, Wayne State University, Detroit, Michigan, United States of America
| | - Sungchil Yang
- Helen Wills Neuroscience Institute, University of California, Berkeley, California, United States of America
- Department of Biomedical Science, City University of Hong Kong, Kowloon, Hong Kong
| | - Shaowen Bao
- Department of Physiology, University of Arizona, Tucson, Arizona, United States of America
- Helen Wills Neuroscience Institute, University of California, Berkeley, California, United States of America
- * E-mail:
| |
Collapse
|
30
|
Auerbach BD, Radziwon K, Salvi R. Testing the Central Gain Model: Loudness Growth Correlates with Central Auditory Gain Enhancement in a Rodent Model of Hyperacusis. Neuroscience 2019; 407:93-107. [PMID: 30292765 PMCID: PMC8792806 DOI: 10.1016/j.neuroscience.2018.09.036] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/18/2018] [Accepted: 09/25/2018] [Indexed: 10/28/2022]
Abstract
The central gain model of hyperacusis proposes that loss of auditory input can result in maladaptive neuronal gain increases in the central auditory system, leading to the over-amplification of sound-evoked activity and excessive loudness perception. Despite the attractiveness of this model, and supporting evidence for it, a critical test of the central gain theory requires that changes in sound-evoked activity be explicitly linked to perceptual alterations of loudness. Here we combined an operant conditioning task that uses a subject's reaction time to auditory stimuli to produce reliable measures of loudness growth with chronic electrophysiological recordings from the auditory cortex and inferior colliculus of awake, behaviorally-phenotyped animals. In this manner, we could directly correlate daily assessments of loudness perception with neurophysiological measures of sound encoding within the same animal. We validated this novel psychophysical-electrophysiological paradigm with a salicylate-induced model of hearing loss and hyperacusis, as high doses of sodium salicylate reliably induce temporary hearing loss, neural hyperactivity, and auditory perceptual disruptions like tinnitus and hyperacusis. Salicylate induced parallel changes to loudness growth and evoked response-intensity functions consistent with temporary hearing loss and hyperacusis. Most importantly, we found that salicylate-mediated changes in loudness growth and sound-evoked activity were correlated within individual animals. These results provide strong support for the central gain model of hyperacusis and demonstrate the utility of using an experimental design that allows for within-subject comparison of behavioral and electrophysiological measures, thereby making inter-subject variability a strength rather than a limitation.
Collapse
Affiliation(s)
- Benjamin D Auerbach
- Center for Hearing and Deafness, State University of New York at Buffalo, Buffalo, NY 14214, USA.
| | - Kelly Radziwon
- Center for Hearing and Deafness, State University of New York at Buffalo, Buffalo, NY 14214, USA
| | - Richard Salvi
- Center for Hearing and Deafness, State University of New York at Buffalo, Buffalo, NY 14214, USA
| |
Collapse
|
31
|
Pienkowski M. Rationale and Efficacy of Sound Therapies for Tinnitus and Hyperacusis. Neuroscience 2019; 407:120-134. [DOI: 10.1016/j.neuroscience.2018.09.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/05/2018] [Accepted: 09/07/2018] [Indexed: 12/20/2022]
|
32
|
Qin D, Liu P, Chen H, Huang X, Ye W, Lin X, Wei F, Su J. Salicylate-Induced Ototoxicity of Spiral Ganglion Neurons: Ca 2+/CaMKII-Mediated Interaction Between NMDA Receptor and GABA A Receptor. Neurotox Res 2019; 35:838-847. [PMID: 30820888 DOI: 10.1007/s12640-019-0006-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/21/2019] [Accepted: 01/24/2019] [Indexed: 10/27/2022]
Abstract
Sodium salicylate (SS) is one of the nonsteroidal anti-inflammatory drugs and widely used in clinical practice. Therefore, we aimed to investigate the potential ototoxicity mechanism of sodium salicylate: the influence of Ca2+/calmodulin-dependent protein kinase II (Ca2+/CaMKII) in interaction between NMDA receptors (NMDAR) and GABAA receptors (GABAAR) in rat cochlear spiral ganglion neurons (SGNs). After treatment with SS, NMDA, and an NMDAR inhibitor (APV), the changes of GABAAR β3 (GABR β3) mRNA, surface and total protein, and GABAAR currents in SGNs were assessed by quantitative PCR, Western blot, and whole-cell patch clamp. Mechanistically, SS and/or NMDA increased the GABR β3 mRNA expression, while decreased GABR β3 surface protein levels and GABAAR-mediated currents. Moreover, application of SS and/or NMDA showed promotion in phosphorylation levels at S383 of GABR β3. Collectively, Ca2+ chelator (BAPTA) or Ca2+/CaMKII inhibitor (KN-93) reversed the effects of SS and/or NMDA on GABAAR. Therefore, we hypothesize that the interaction between NMDAR and GABAAR is involved in the SGNs damage induced by SS. In addition, the underlying molecular mechanism is related to Ca2+/CaMKII-mediated signaling pathway, which suggests that the interaction between calcium signal-regulated receptors mediates SS ototoxicity.
Collapse
Affiliation(s)
- Danxue Qin
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Peiqiang Liu
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Huiying Chen
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xi Huang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.,Department of Otolaryngology-Head and Neck Surgery, Wuhan No.1 Hospital, Wuhan, 430022, Hubei, China
| | - Wenhua Ye
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiaoyu Lin
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Fangyu Wei
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jiping Su
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
33
|
Balaram P, Hackett TA, Polley DB. Synergistic Transcriptional Changes in AMPA and GABA A Receptor Genes Support Compensatory Plasticity Following Unilateral Hearing Loss. Neuroscience 2018; 407:108-119. [PMID: 30176318 DOI: 10.1016/j.neuroscience.2018.08.023] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 08/02/2018] [Accepted: 08/22/2018] [Indexed: 01/11/2023]
Abstract
Debilitating perceptual disorders including tinnitus, hyperacusis, phantom limb pain and visual release hallucinations may reflect aberrant patterns of neural activity in central sensory pathways following a loss of peripheral sensory input. Here, we explore short- and long-term changes in gene expression that may contribute to hyperexcitability following a sudden, profound loss of auditory input from one ear. We used fluorescence in situ hybridization to quantify mRNA levels for genes encoding AMPA and GABAA receptor subunits (Gria2 and Gabra1, respectively) in single neurons from the inferior colliculus (IC) and auditory cortex (ACtx). Thirty days after unilateral hearing loss, Gria2 levels were significantly increased while Gabra1 levels were significantly decreased. Transcriptional rebalancing was more pronounced in ACtx than IC and bore no obvious relationship to the degree of hearing loss. By contrast to the opposing, synergistic shifts in Gria2 and Gabra1 observed 30 days after hearing loss, we found that transcription levels for both genes were equivalently reduced after 5 days of hearing loss, producing no net change in the excitatory/inhibitory transcriptional balance. Opposing transcriptional shifts in AMPA and GABA receptor genes that emerge several weeks after a peripheral insult could promote both sensitization and disinhibition to support a homeostatic recovery of neural activity following auditory deprivation. Imprecise transcriptional changes could also drive the system toward perceptual hypersensitivity, degraded temporal processing and the irrepressible perception of non-existent environmental stimuli, a trio of perceptual impairments that often accompany chronic sensory deprivation.
Collapse
Affiliation(s)
- P Balaram
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear Infirmary, Boston MA 02114, USA; Dept. of Otolaryngology, Harvard Medical School, Boston MA 02114, USA
| | - T A Hackett
- Dept. of Hearing and Speech Sciences, Vanderbilt Bill Wilkerson Center for Otolaryngology and Communication Sciences, Vanderbilt University Medical Center, Nashville TN 37232 USA
| | - D B Polley
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear Infirmary, Boston MA 02114, USA; Dept. of Otolaryngology, Harvard Medical School, Boston MA 02114, USA.
| |
Collapse
|
34
|
Central Compensation in Auditory Brainstem after Damaging Noise Exposure. eNeuro 2018; 5:eN-CFN-0250-18. [PMID: 30123822 PMCID: PMC6096756 DOI: 10.1523/eneuro.0250-18.2018] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 07/19/2018] [Indexed: 12/15/2022] Open
Abstract
Noise exposure is one of the most common causes of hearing loss and peripheral damage to the auditory system. A growing literature suggests that the auditory system can compensate for peripheral loss through increased central neural activity. The current study sought to investigate the link between noise exposure, increases in central gain, synaptic reorganization, and auditory function. All axons of the auditory nerve project to the cochlear nucleus, making it a requisite nucleus for sound detection. As the first synapse in the central auditory system, the cochlear nucleus is well positioned to respond plastically to loss of peripheral input. To investigate noise-induced compensation in the central auditory system, we measured auditory brainstem responses (ABRs) and auditory perception and collected tissue from mice exposed to broadband noise. Noise-exposed mice showed elevated ABR thresholds, reduced ABR wave 1 amplitudes, and spiral ganglion neuron loss. Despite peripheral damage, noise-exposed mice were hyperreactive to loud sounds and showed nearly normal behavioral sound detection thresholds. Ratios of late ABR peaks (2–4) relative to the first ABR peak indicated that brainstem pathways were hyperactive in noise-exposed mice, while anatomical analysis indicated there was an imbalance between expression of excitatory and inhibitory proteins in the ventral cochlear nucleus. The results of the current study suggest that a reorganization of excitation and inhibition in the ventral cochlear nucleus may drive hyperactivity in the central auditory system. This increase in central gain can compensate for peripheral loss to restore some aspects of auditory function.
Collapse
|
35
|
Baizer JS, Wong KM, Salvi RJ, Manohar S, Sherwood CC, Hof PR, Baker JF, Witelson SF. Species Differences in the Organization of the Ventral Cochlear Nucleus. Anat Rec (Hoboken) 2018; 301:862-886. [PMID: 29236365 PMCID: PMC5902649 DOI: 10.1002/ar.23751] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 08/18/2017] [Accepted: 09/17/2017] [Indexed: 01/18/2023]
Abstract
The mammalian cochlear nuclei (CN) consist of two major subdivisions, the dorsal (DCN) and ventral (VCN) nuclei. We previously reported differences in the structural and neurochemical organization of the human DCN from that in several other species. Here we extend this analysis to the VCN, considering both the organization of subdivisions and the types and distributions of neurons. Classically, the VCN in mammals is composed of two subdivisions, the anteroventral (VCA) and posteroventral cochlear nuclei (VCP). Anatomical and electrophysiological data in several species have defined distinct neuronal types with different distributions in the VCA and VCP. We asked if VCN subdivisions and anatomically defined neuronal types might be distinguished by patterns of protein expression in humans. We also asked if the neurochemical characteristics of the VCN are the same in humans as in other mammalian species, analyzing data from chimpanzees, macaque monkeys, cats, rats and chinchillas. We examined Nissl- and immunostained sections, using antibodies that had labeled neurons in other brainstem nuclei in humans. Nissl-stained sections supported the presence of both VCP and VCA in humans and chimpanzees. However, patterns of protein expression did not differentiate classes of neurons in humans; neurons of different soma shapes and dendritic configurations all expressed the same proteins. The patterns of immunostaining in macaque monkey, cat, rat, and chinchilla were different from those in humans and chimpanzees and from each other. The results may correlate with species differences in auditory function and plasticity. Anat Rec, 301:862-886, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Joan S Baizer
- Department of Physiology and Biophysics, University at Buffalo, Buffalo, New York
| | - Keit Men Wong
- Department of Physiology and Biophysics, University at Buffalo, Buffalo, New York
| | - Richard J Salvi
- Department of Communicative Disorders and Sciences, Center for Hearing and Deafness, University at Buffalo, Buffalo, New York
| | - Senthilvelan Manohar
- Department of Communicative Disorders and Sciences, Center for Hearing and Deafness, University at Buffalo, Buffalo, New York
| | - Chet C Sherwood
- Department of Anthropology, The George Washington University, Washington, DC
| | - Patrick R Hof
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - James F Baker
- Department of Physiology, Northwestern University Medical School, Chicago, Illinois
| | - Sandra F Witelson
- Department of Psychiatry and Behavioural Neurosciences, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, L8S 4K1, Canada
| |
Collapse
|
36
|
Ruan Q, Yu Z, Zhang W, Ruan J, Liu C, Zhang R. Cholinergic Hypofunction in Presbycusis-Related Tinnitus With Cognitive Function Impairment: Emerging Hypotheses. Front Aging Neurosci 2018; 10:98. [PMID: 29681847 PMCID: PMC5897739 DOI: 10.3389/fnagi.2018.00098] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 03/22/2018] [Indexed: 01/21/2023] Open
Abstract
Presbycusis (age-related hearing loss) is a potential risk factor for tinnitus and cognitive deterioration, which result in poor life quality. Presbycusis-related tinnitus with cognitive impairment is a common phenotype in the elderly population. In these individuals, the central auditory system shows similar pathophysiological alterations as those observed in Alzheimer's disease (AD), including cholinergic hypofunction, epileptiform-like network synchronization, chronic inflammation, and reduced GABAergic inhibition and neural plasticity. Observations from experimental rodent models indicate that recovery of cholinergic function can improve memory and other cognitive functions via acetylcholine-mediated GABAergic inhibition enhancement, nicotinic acetylcholine receptor (nAChR)-mediated anti-inflammation, glial activation inhibition and neurovascular protection. The loss of cholinergic innervation of various brain structures may provide a common link between tinnitus seen in presbycusis-related tinnitus and age-related cognitive impairment. We hypothesize a key component of the condition is the withdrawal of cholinergic input to a subtype of GABAergic inhibitory interneuron, neuropeptide Y (NPY) neurogliaform cells. Cholinergic denervation might not only cause the degeneration of NPY neurogliaform cells, but may also result in decreased AChR activation in GABAergic inhibitory interneurons. This, in turn, would lead to reduced GABA release and inhibitory regulation of neural networks. Reduced nAChR-mediated anti-inflammation due to the loss of nicotinic innervation might lead to the transformation of glial cells and release of inflammatory mediators, lowering the buffering of extracellular potassium and glutamate metabolism. Further research will provide evidence for the recovery of cholinergic function with the use of cholinergic input enhancement alone or in combination with other rehabilitative interventions to reestablish inhibitory regulation mechanisms of involved neural networks for presbycusis-related tinnitus with cognitive impairment.
Collapse
Affiliation(s)
- Qingwei Ruan
- Shanghai Institute of Geriatrics and Gerontology, Shanghai Key Laboratory of Clinical Geriatrics, Huadong Hospital, and Research Center of Aging and Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhuowei Yu
- Shanghai Institute of Geriatrics and Gerontology, Shanghai Key Laboratory of Clinical Geriatrics, Huadong Hospital, and Research Center of Aging and Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weibin Zhang
- Shanghai Institute of Geriatrics and Gerontology, Shanghai Key Laboratory of Clinical Geriatrics, Huadong Hospital, and Research Center of Aging and Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian Ruan
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunhui Liu
- Department of Otolaryngology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ruxin Zhang
- Department of Otolaryngology, Huadong Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
37
|
Gauvin DV, Yoder J, Zimmermann ZJ, Tapp R. Ototoxicity: The Radical Drum Beat and Rhythm of Cochlear Hair Cell Life and Death. Int J Toxicol 2018; 37:195-206. [PMID: 29575954 DOI: 10.1177/1091581818761128] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The function and structure of the auditory information processing system establishes a unique sensory environment for the "perfect storm." The battle between life and death pits the cascade of an apoptotic storm, programmed cell death cascades, against simple cell death (necrosis) pathways. Live or die, the free radical biology of oxygen and hydroxylation, and the destruction of transition metal migration through the mechanical gate sensory processes of the hair cell lead to direct access to the cytoplasm, cytoplasmic reticulum, and mitochondria of the inner workings of the hair cells. These lead to subsequent interactions with nuclear DNA resulting in permanent hearing loss. The yin and yang of pharmaceutical product development is to document what kills, why it kills, and how do we mitigate it. This review highlights the processes of cell death within the cochlea.
Collapse
Affiliation(s)
- David V Gauvin
- 1 Neurobehavioral Sciences Department, MPI Research, Inc., Mattawan, MI, USA
| | - Joshua Yoder
- 1 Neurobehavioral Sciences Department, MPI Research, Inc., Mattawan, MI, USA
| | | | - Rachel Tapp
- 1 Neurobehavioral Sciences Department, MPI Research, Inc., Mattawan, MI, USA
| |
Collapse
|
38
|
Acoustic Trauma Changes the Parvalbumin-Positive Neurons in Rat Auditory Cortex. Neural Plast 2018; 2018:9828070. [PMID: 29593786 PMCID: PMC5822889 DOI: 10.1155/2018/9828070] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 10/03/2017] [Accepted: 11/26/2017] [Indexed: 11/18/2022] Open
Abstract
Acoustic trauma is being reported to damage the auditory periphery and central system, and the compromised cortical inhibition is involved in auditory disorders, such as hyperacusis and tinnitus. Parvalbumin-containing neurons (PV neurons), a subset of GABAergic neurons, greatly shape and synchronize neural network activities. However, the change of PV neurons following acoustic trauma remains to be elucidated. The present study investigated how auditory cortical PV neurons change following unilateral 1 hour noise exposure (left ear, one octave band noise centered at 16 kHz, 116 dB SPL). Noise exposure elevated the auditory brainstem response threshold of the exposed ear when examined 7 days later. More detectable PV neurons were observed in both sides of the auditory cortex of noise-exposed rats when compared to control. The detectable PV neurons of the left auditory cortex (ipsilateral to the exposed ear) to noise exposure outnumbered those of the right auditory cortex (contralateral to the exposed ear). Quantification of Western blotted bands revealed higher expression level of PV protein in the left cortex. These findings of more active PV neurons in noise-exposed rats suggested that a compensatory mechanism might be initiated to maintain a stable state of the brain.
Collapse
|
39
|
Sottile SY, Ling L, Cox BC, Caspary DM. Impact of ageing on postsynaptic neuronal nicotinic neurotransmission in auditory thalamus. J Physiol 2017; 595:5375-5385. [PMID: 28585699 PMCID: PMC5538226 DOI: 10.1113/jp274467] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 05/24/2017] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Neuronal nicotinic acetylcholine receptors (nAChRs) play a fundamental role in the attentional circuitry throughout the mammalian CNS. In the present study, we report a novel finding that ageing negatively impacts nAChR efficacy in auditory thalamus, and this is probably the result of a loss of nAChR density (Bmax ) and changes in the subunit composition of nAChRs. Our data support the hypothesis that age-related maladaptive changes involving nAChRs within thalamocortical circuits partially underpin the difficulty that elderly adults experience with respect to attending to speech and other salient acoustic signals. ABSTRACT The flow of auditory information through the medial geniculate body (MGB) is regulated, in part, by cholinergic projections from the pontomesencephalic tegmentum. The functional significance of these projections is not fully established, although they have been strongly implicated in the allocation of auditory attention. Using in vitro slice recordings, we have analysed postsynaptic function and pharmacology of neuronal nicotinic ACh receptors (nAChRs) in young adult and the aged rat MGB. We find that ACh produces significant excitatory postsynaptic actions on young MGB neurons, probably mediated by β2-containing heteromeric nAChRs. Radioligand binding studies show a significant age-related loss of heteromeric nAChR receptor number, which supports patch clamp data showing an age-related loss in ACh efficacy in evoking postsynaptic responses. Use of the β2-selective nAChR antagonist, dihydro-β-erythroidine, suggests that loss of cholinergic efficacy may also be the result of an age-related subunit switch from high affinity β2-containing nAChRs to low affinity β4-containing nAChRs, in addition to the loss of total nAChR number. This age-related nAChR dysfunction may partially underpin the attentional deficits that contribute to the loss of speech understanding in the elderly.
Collapse
Affiliation(s)
| | | | - Brandon C. Cox
- Department of Pharmacology
- Department of Surgery, Division of OtolaryngologySouthern Illinois University School of MedicineSpringfieldILUSA
| | - Donald M. Caspary
- Department of Pharmacology
- Department of Surgery, Division of OtolaryngologySouthern Illinois University School of MedicineSpringfieldILUSA
| |
Collapse
|
40
|
Noise Trauma-Induced Behavioral Gap Detection Deficits Correlate with Reorganization of Excitatory and Inhibitory Local Circuits in the Inferior Colliculus and Are Prevented by Acoustic Enrichment. J Neurosci 2017; 37:6314-6330. [PMID: 28583912 DOI: 10.1523/jneurosci.0602-17.2017] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 05/04/2017] [Accepted: 05/14/2017] [Indexed: 01/12/2023] Open
Abstract
Hearing loss leads to a host of cellular and synaptic changes in auditory brain areas that are thought to give rise to auditory perception deficits such as temporal processing impairments, hyperacusis, and tinnitus. However, little is known about possible changes in synaptic circuit connectivity that may underlie these hearing deficits. Here, we show that mild hearing loss as a result of brief noise exposure leads to a pronounced reorganization of local excitatory and inhibitory circuits in the mouse inferior colliculus. The exact nature of these reorganizations correlated with the presence or absence of the animals' impairments in detecting brief sound gaps, a commonly used behavioral sign for tinnitus in animal models. Mice with gap detection deficits (GDDs) showed a shift in the balance of synaptic excitation and inhibition that was present in both glutamatergic and GABAergic neurons, whereas mice without GDDs showed stable excitation-inhibition balances. Acoustic enrichment (AE) with moderate intensity, pulsed white noise immediately after noise trauma prevented both circuit reorganization and GDDs, raising the possibility of using AE immediately after cochlear damage to prevent or alleviate the emergence of central auditory processing deficits.SIGNIFICANCE STATEMENT Noise overexposure is a major cause of central auditory processing disorders, including tinnitus, yet the changes in synaptic connectivity underlying these disorders remain poorly understood. Here, we find that brief noise overexposure leads to distinct reorganizations of excitatory and inhibitory synaptic inputs onto glutamatergic and GABAergic neurons and that the nature of these reorganizations correlates with animals' impairments in detecting brief sound gaps, which is often considered a sign of tinnitus. Acoustic enrichment immediately after noise trauma prevents circuit reorganizations and gap detection deficits, highlighting the potential for using sound therapy soon after cochlear damage to prevent the development of central processing deficits.
Collapse
|
41
|
Brecht EJ, Barsz K, Gross B, Walton JP. Increasing GABA reverses age-related alterations in excitatory receptive fields and intensity coding of auditory midbrain neurons in aged mice. Neurobiol Aging 2017; 56:87-99. [PMID: 28532644 DOI: 10.1016/j.neurobiolaging.2017.04.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 03/18/2017] [Accepted: 04/04/2017] [Indexed: 11/25/2022]
Abstract
A key feature of age-related hearing loss is a reduction in the expression of inhibitory neurotransmitters in the central auditory system. This loss is partially responsible for changes in central auditory processing, as inhibitory receptive fields play a critical role in shaping neural responses to sound stimuli. Vigabatrin (VGB), an antiepileptic agent that irreversibly inhibits γ-amino butyric acid (GABA) transaminase, leads to increased availability of GABA throughout the brain. This study used multi-channel electrophysiology measurements to assess the excitatory frequency response areas in old CBA mice to which VGB had been administered. We found a significant post-VGB reduction in the proportion of V-type shapes, and an increase in primary-like excitatory frequency response areas. There was also a significant increase in the mean maximum driven spike rates across the tonotopic frequency range of all treated animals, consistent with observations that GABA buildup within the central auditory system increases spike counts of neural receptive fields. This increased spiking is also seen in the rate-level functions and seems to explain the improved low-frequency thresholds.
Collapse
Affiliation(s)
- Elliott J Brecht
- Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL, USA; Global Center of Speech and Hearing Research, University of South Florida, Tampa, FL, USA
| | - Kathy Barsz
- School of Nursing, University of Rochester, Rochester, NY, USA
| | - Benjamin Gross
- Global Center of Speech and Hearing Research, University of South Florida, Tampa, FL, USA; Department of Physics, University of South Florida, Tampa, FL, USA
| | - Joseph P Walton
- Department of Chemical and Biomedical Engineering, University of South Florida, Tampa, FL, USA; Global Center of Speech and Hearing Research, University of South Florida, Tampa, FL, USA; Department of Communication Sciences and Disorders, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
42
|
Jiang C, Luo B, Manohar S, Chen GD, Salvi R. Plastic changes along auditory pathway during salicylate-induced ototoxicity: Hyperactivity and CF shifts. Hear Res 2017; 347:28-40. [PMID: 27989950 PMCID: PMC5403591 DOI: 10.1016/j.heares.2016.10.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 09/30/2016] [Accepted: 10/26/2016] [Indexed: 12/27/2022]
Abstract
High dose of salicylate, the active ingredient in aspirin, has long been known to induce transient hearing loss, tinnitus and hyperacusis making it a powerful experimental tool. These salicylate-induced perceptual disturbances are associated with a massive reduction in the neural output of the cochlea. Paradoxically, the diminished neural output of the cochlea is accompanied by a dramatic increase in sound-evoked activity in the auditory cortex (AC) and several other parts of the central nervous system. Exactly where the increase in neural activity begins and builds up along the central auditory pathway are not fully understood. To address this issue, we measured sound-evoked neural activity in the cochlea, cochlear nucleus (CN), inferior colliculus (IC), and AC before and after administering a high dose of sodium salicylate (SS, 300 mg/kg). The SS-treatment abolished low-level sound-evoked responses along the auditory pathway resulting in a 20-30 dB threshold shift. While the neural output of the cochlea was substantially reduced at high intensities, the neural responses in the CN were only slightly reduced; those in the IC were nearly normal or slightly enhanced while those in the AC considerably enhanced, indicative of a progress increase in central gain. The SS-induced increase in central response in the IC and AC was frequency-dependent with the greatest increase occurring in the mid-frequency range the putative pitch of SS-induced tinnitus. This frequency-dependent hyperactivity appeared to result from shifts in the frequency receptive fields (FRF) such that the response areas of many FRF shifted/expanded toward the mid-frequencies. Our results suggest that the SS-induced threshold shift originates in the cochlea. In contrast, enhanced central gain is not localized to one region, but progressively builds up at successively higher stage of the auditory pathway either through a loss of inhibition and/or increased excitation.
Collapse
Affiliation(s)
- Chen Jiang
- Department of Neurosurgery, Anhui Provincial Hospital, 17 Lujiang Road, Hefei, Anhui 230001, China; Center for Hearing and Deafness, State University of New York at Buffalo, 137 Cary Hall, 3435 Main Street, Buffalo, NY 14214, USA
| | - Bin Luo
- Department of Neurosurgery, Anhui Provincial Hospital, 17 Lujiang Road, Hefei, Anhui 230001, China; Center for Hearing and Deafness, State University of New York at Buffalo, 137 Cary Hall, 3435 Main Street, Buffalo, NY 14214, USA
| | - Senthilvelan Manohar
- Center for Hearing and Deafness, State University of New York at Buffalo, 137 Cary Hall, 3435 Main Street, Buffalo, NY 14214, USA
| | - Guang-Di Chen
- Center for Hearing and Deafness, State University of New York at Buffalo, 137 Cary Hall, 3435 Main Street, Buffalo, NY 14214, USA.
| | - Richard Salvi
- Center for Hearing and Deafness, State University of New York at Buffalo, 137 Cary Hall, 3435 Main Street, Buffalo, NY 14214, USA
| |
Collapse
|
43
|
He J, Zhu Y, Aa J, Smith PF, De Ridder D, Wang G, Zheng Y. Brain Metabolic Changes in Rats following Acoustic Trauma. Front Neurosci 2017; 11:148. [PMID: 28392756 PMCID: PMC5364180 DOI: 10.3389/fnins.2017.00148] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 03/09/2017] [Indexed: 12/14/2022] Open
Abstract
Acoustic trauma is the most common cause of hearing loss and tinnitus in humans. However, the impact of acoustic trauma on system biology is not fully understood. It has been increasingly recognized that tinnitus caused by acoustic trauma is unlikely to be generated by a single pathological source, but rather a complex network of changes involving not only the auditory system but also systems related to memory, emotion and stress. One obvious and significant gap in tinnitus research is a lack of biomarkers that reflect the consequences of this interactive "tinnitus-causing" network. In this study, we made the first attempt to analyse brain metabolic changes in rats following acoustic trauma using metabolomics, as a pilot study prior to directly linking metabolic changes to tinnitus. Metabolites in 12 different brain regions collected from either sham or acoustic trauma animals were profiled using a gas chromatography mass spectrometry (GC/MS)-based metabolomics platform. After deconvolution of mass spectra and identification of the molecules, the metabolomic data were processed using multivariate statistical analysis. Principal component analysis showed that metabolic patterns varied among different brain regions; however, brain regions with similar functions had a similar metabolite composition. Acoustic trauma did not change the metabolite clusters in these regions. When analyzed within each brain region using the orthogonal projection to latent structures discriminant analysis sub-model, 17 molecules showed distinct separation between control and acoustic trauma groups in the auditory cortex, inferior colliculus, superior colliculus, vestibular nucleus complex (VNC), and cerebellum. Further metabolic pathway impact analysis and the enrichment overview with network analysis suggested the primary involvement of amino acid metabolism, including the alanine, aspartate and glutamate metabolic pathways, the arginine and proline metabolic pathways and the purine metabolic pathway. Our results provide the first metabolomics evidence that acoustic trauma can induce changes in multiple metabolic pathways. This pilot study also suggests that the metabolomic approach has the potential to identify acoustic trauma-specific metabolic shifts in future studies where metabolic changes are correlated with the animal's tinnitus status.
Collapse
Affiliation(s)
- Jun He
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University Nanjing, Jiangsu, China
| | - Yejin Zhu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University Nanjing, Jiangsu, China
| | - Jiye Aa
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University Nanjing, Jiangsu, China
| | - Paul F Smith
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of OtagoDunedin, New Zealand; Brain Health Research Centre, University of OtagoDunedin, New Zealand; Brain Research New ZealandDunedin, New Zealand; Eisdell Moore Centre for Hearing and Balance Research, University of AucklandAuckland, New Zealand
| | - Dirk De Ridder
- Brain Health Research Centre, University of OtagoDunedin, New Zealand; Brain Research New ZealandDunedin, New Zealand; Eisdell Moore Centre for Hearing and Balance Research, University of AucklandAuckland, New Zealand; Department of Neurosurgery, Dunedin Medical School, University of OtagoOtago, New Zealand
| | - Guangji Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University Nanjing, Jiangsu, China
| | - Yiwen Zheng
- Department of Pharmacology and Toxicology, School of Biomedical Sciences, University of OtagoDunedin, New Zealand; Brain Health Research Centre, University of OtagoDunedin, New Zealand; Brain Research New ZealandDunedin, New Zealand; Eisdell Moore Centre for Hearing and Balance Research, University of AucklandAuckland, New Zealand
| |
Collapse
|
44
|
Salvi R, Sun W, Ding D, Chen GD, Lobarinas E, Wang J, Radziwon K, Auerbach BD. Inner Hair Cell Loss Disrupts Hearing and Cochlear Function Leading to Sensory Deprivation and Enhanced Central Auditory Gain. Front Neurosci 2017; 10:621. [PMID: 28149271 PMCID: PMC5241314 DOI: 10.3389/fnins.2016.00621] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 12/30/2016] [Indexed: 11/13/2022] Open
Abstract
There are three times as many outer hair cells (OHC) as inner hair cells (IHC), yet IHC transmit virtually all acoustic information to the brain as they synapse with 90–95% of type I auditory nerve fibers. Here we review a comprehensive series of experiments aimed at determining how loss of the IHC/type I system affects hearing by selectively destroying these cells in chinchillas using the ototoxic anti-cancer agent carboplatin. Eliminating IHC/type I neurons has no effect on distortion product otoacoustic emission or the cochlear microphonic potential generated by OHC; however, it greatly reduces the summating potential produced by IHC and the compound action potential (CAP) generated by type I neurons. Remarkably, responses from remaining auditory nerve fibers maintain sharp tuning and low thresholds despite innervating regions of the cochlea with ~80% IHC loss. Moreover, chinchillas with large IHC lesions have surprisingly normal thresholds in quiet until IHC losses exceeded 80%, suggesting that only a few IHC are needed to detect sounds in quiet. However, behavioral thresholds in broadband noise are elevated significantly and tone-in-narrow band noise masking patterns exhibit greater remote masking. These results suggest the auditory system is able to compensate for considerable loss of IHC/type I neurons in quiet but not in difficult listening conditions. How does the auditory brain deal with the drastic loss of cochlear input? Recordings from the inferior colliculus found a relatively small decline in sound-evoked activity despite a large decrease in CAP amplitude after IHC lesion. Paradoxically, sound-evoked responses are generally larger than normal in the auditory cortex, indicative of increased central gain. This gain enhancement in the auditory cortex is associated with decreased GABA-mediated inhibition. These results suggest that when the neural output of the cochlea is reduced, the central auditory system compensates by turning up its gain so that weak signals once again become comfortably loud. While this gain enhancement is able to restore normal hearing under quiet conditions, it may not adequately compensate for peripheral dysfunction in more complex sound environments. In addition, excessive gain increases may convert recruitment into the debilitating condition known as hyperacusis.
Collapse
Affiliation(s)
- Richard Salvi
- Center for Hearing and Deafness, University at Buffalo Buffalo, NY, USA
| | - Wei Sun
- Center for Hearing and Deafness, University at Buffalo Buffalo, NY, USA
| | - Dalian Ding
- Center for Hearing and Deafness, University at Buffalo Buffalo, NY, USA
| | - Guang-Di Chen
- Center for Hearing and Deafness, University at Buffalo Buffalo, NY, USA
| | | | - Jian Wang
- School of Human Communication Disorders, Dalhousie University Halifax, NS, Canada
| | - Kelly Radziwon
- Center for Hearing and Deafness, University at Buffalo Buffalo, NY, USA
| | | |
Collapse
|
45
|
Acquired hearing loss and brain plasticity. Hear Res 2017; 343:176-190. [DOI: 10.1016/j.heares.2016.05.008] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 04/29/2016] [Accepted: 05/19/2016] [Indexed: 12/19/2022]
|
46
|
Kapolowicz MR, Thompson LT. Acute high-intensity noise induces rapid Arc protein expression but fails to rapidly change GAD expression in amygdala and hippocampus of rats: Effects of treatment with D-cycloserine. Hear Res 2016; 342:69-79. [PMID: 27702572 DOI: 10.1016/j.heares.2016.09.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 08/26/2016] [Accepted: 09/30/2016] [Indexed: 10/20/2022]
Abstract
Tinnitus is a devastating auditory disorder impacting a growing number of people each year. The aims of the current experiment were to assess neuronal mechanisms involved in the initial plasticity after traumatic noise exposure that could contribute to the emergence of tinnitus and to test a potential pharmacological treatment to alter this early neural plasticity. Specifically, this study addressed rapid effects of acute noise trauma on amygdalo-hippocampal circuitry, characterizing biomarkers of both excitation and inhibition in these limbic regions, and compared them to expression of these same markers in primary auditory cortex shortly after acute noise trauma. To assess excitatory plasticity, activity-regulated cytoskeleton-associated (Arc) protein expression was evaluated in male rats 45 min after bilateral exposure to acute high-intensity noise (16 kHz, 115 dB SPL, for 1 h), sufficient to cause acute cochlear trauma, a common cause of tinnitus in humans and previously shown sufficient to induce tinnitus in rat models of this auditory neuropathology. Western blot analyses confirmed that up-regulation of amygdalo-hippocampal Arc expression occurred rapidly post-noise trauma, corroborating several lines of evidence from our own and other laboratories indicating that limbic brain structures, i.e. outside of the classical auditory pathways, exhibit plasticity early in the initiation of tinnitus. Western blot analyses revealed no noise-induced changes in amygdalo-hippocampal expression of glutamate decarboxylase (GAD), the biosynthetic enzyme required for GABAergic inhibition. No changes in either Arc or GAD protein expression were observed in primary auditory cortex in this immediate post-noise exposure period, confirming other reports that auditory cortical plasticity may not occur until later in the development of tinnitus. As a further control, our experiments compared Arc protein expression between groups exposed to the quiet background of a sound-proof chamber to those exposed not only to the traumatic noise described above, but also to an intermediate, non-traumatic noise level (70 dB SPL) for the same duration in each of these three brain regions. We found that non-traumatic noise did not up-regulate Arc protein expression in these brain regions. To see if changes in Arc expression due to acute traumatic noise exposure were stress-related, we compared circulating serum corticosterone in controls and rats exposed to traumatic noise at the time when changes in Arc were observed, and found no significant differences in this stress hormone in our experimental conditions. Finally, the ability of D-cycloserine (DCS; an NMDA-receptor NR1 partial agonist) to reduce or prevent the noise trauma-related plastic changes in the biomarker, Arc, was tested. D-cycloserine prevented traumatic noise-induced up-regulation of Arc protein expression in amygdala but not in hippocampus, suggesting that DCS alone is not fully effective in eliminating regionally-specific early plastic changes after traumatic noise exposure.
Collapse
Affiliation(s)
- M R Kapolowicz
- Behavioral & Brain Sciences, Neuroscience, The University of Texas at Dallas, 800W. Campbell Rd., BSB 14, Richardson, TX, 75080, USA
| | - L T Thompson
- Behavioral & Brain Sciences, Neuroscience, The University of Texas at Dallas, 800W. Campbell Rd., BSB 14, Richardson, TX, 75080, USA.
| |
Collapse
|
47
|
Tighilet B, Dutheil S, Siponen MI, Noreña AJ. Reactive Neurogenesis and Down-Regulation of the Potassium-Chloride Cotransporter KCC2 in the Cochlear Nuclei after Cochlear Deafferentation. Front Pharmacol 2016; 7:281. [PMID: 27630564 PMCID: PMC5005331 DOI: 10.3389/fphar.2016.00281] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 08/16/2016] [Indexed: 12/26/2022] Open
Abstract
While many studies have been devoted to investigating the homeostatic plasticity triggered by cochlear hearing loss, the cellular and molecular mechanisms involved in these central changes remain elusive. In the present study, we investigated the possibility of reactive neurogenesis after unilateral cochlear nerve section in the cochlear nucleus (CN) of cats. We found a strong cell proliferation in all the CN sub-divisions ipsilateral to the lesion. Most of the newly generated cells survive up to 1 month after cochlear deafferentation in all cochlear nuclei (except the dorsal CN) and give rise to a variety of cell types, i.e., microglial cells, astrocytes, and neurons. Interestingly, many of the newborn neurons had an inhibitory (GABAergic) phenotype. This result is intriguing since sensory deafferentation is usually accompanied by enhanced excitation, consistent with a reduction in central inhibition. The membrane potential effect of GABA depends, however, on the intra-cellular chloride concentration, which is maintained at low levels in adults by the potassium chloride co-transporter KCC2. The KCC2 density on the plasma membrane of neurons was then assessed after cochlear deafferentation in the cochlear nuclei ipsilateral and contralateral to the lesion. Cochlear deafferentation is accompanied by a strong down-regulation of KCC2 ipsilateral to the lesion at 3 and 30 days post-lesion. This study suggests that reactive neurogenesis and down-regulation of KCC2 is part of the vast repertoire involved in homeostatic plasticity triggered by hearing loss. These central changes may also play a role in the generation of tinnitus and hyperacusis.
Collapse
Affiliation(s)
- Brahim Tighilet
- Laboratoire de Neurosciences Intégratives et Adaptatives, UMR 7260 - Comportement, Cerveau, Cognition (Behavior, Brain, and Cognition) - Aix-Marseille Université - Centre National de la Recherche Scientifique Marseille, France
| | - Sophie Dutheil
- Department of Psychiatry, School of Medicine, Yale University, New Haven CT, USA
| | - Marina I Siponen
- Laboratoire de Neurosciences Intégratives et Adaptatives, UMR 7260 - Comportement, Cerveau, Cognition (Behavior, Brain, and Cognition) - Aix-Marseille Université - Centre National de la Recherche Scientifique Marseille, France
| | - Arnaud J Noreña
- Laboratoire de Neurosciences Intégratives et Adaptatives, UMR 7260 - Comportement, Cerveau, Cognition (Behavior, Brain, and Cognition) - Aix-Marseille Université - Centre National de la Recherche Scientifique Marseille, France
| |
Collapse
|
48
|
Gao Y, Manzoor N, Kaltenbach JA. Evidence of activity-dependent plasticity in the dorsal cochlear nucleus, in vivo, induced by brief sound exposure. Hear Res 2016; 341:31-42. [PMID: 27490001 DOI: 10.1016/j.heares.2016.07.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 06/27/2016] [Accepted: 07/24/2016] [Indexed: 10/21/2022]
Abstract
The purpose of the present study was to investigate the immediate effects of acute exposure to intense sound on spontaneous and stimulus-driven activity in the dorsal cochlear nucleus (DCN). We examined the levels of multi- and single-unit spontaneous activity before and immediately following brief exposure (2 min) to tones at levels of either 109 or 85 dB SPL. Exposure frequency was selected to either correspond to the units' best frequency (BF) or fall within the borders of its inhibitory side band. The results demonstrate that these exposure conditions caused significant alterations in spontaneous activity and responses to BF tones. The induced changes have a fast onset (minutes) and are persistent for durations of at least 20 min. The directions of the change were found to depend on the frequency of exposure relative to BF. Transient decreases followed by more sustained increases in spontaneous activity were induced when the exposure frequency was at or near the units' BF, while sustained decreases of activity resulted when the exposure frequency fell inside the inhibitory side band. Follow-up studies at the single unit level revealed that the observed activity changes were found on unit types having properties which have previously been found to represent fusiform cells. The changes in spontaneous activity occurred despite only minor changes in response thresholds. Noteworthy changes also occurred in the strength of responses to BF tones, although these changes tended to be in the direction opposite those of the spontaneous rate changes. We discuss the possible role of activity-dependent plasticity as a mechanism underlying the rapid emergence of increased spontaneous activity after tone exposure and suggest that these changes may represent a neural correlate of acute noise-induced tinnitus.
Collapse
Affiliation(s)
- Y Gao
- Department of Neurosciences, Lerner Research Institute, Head and Neck Institute, The Cleveland Clinic, Cleveland, OH, USA
| | - N Manzoor
- Department of Neurosciences, Lerner Research Institute, Head and Neck Institute, The Cleveland Clinic, Cleveland, OH, USA
| | - J A Kaltenbach
- Department of Neurosciences, Lerner Research Institute, Head and Neck Institute, The Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
49
|
Manohar S, Dahar K, Adler HJ, Dalian D, Salvi R. Noise-induced hearing loss: Neuropathic pain via Ntrk1 signaling. Mol Cell Neurosci 2016; 75:101-12. [PMID: 27473923 DOI: 10.1016/j.mcn.2016.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 06/30/2016] [Accepted: 07/25/2016] [Indexed: 12/18/2022] Open
Abstract
Severe noise-induced damage to the inner ear leads to auditory nerve fiber degeneration thereby reducing the neural input to the cochlear nucleus (CN). Paradoxically, this leads to a significant increase in spontaneous activity in the CN which has been linked to tinnitus, hyperacusis and ear pain. The biological mechanisms that lead to an increased spontaneous activity are largely unknown, but could arise from changes in glutamatergic or GABAergic neurotransmission or neuroinflammation. To test this hypothesis, we unilaterally exposed rats for 2h to a 126dB SPL narrow band noise centered at 12kHz. Hearing loss measured by auditory brainstem responses exceeded 55dB from 6 to 32kHz. The mRNA from the exposed CN was harvested at 14 or 28days post-exposure and qRT-PCR analysis was performed on 168 genes involved in neural inflammation, neuropathic pain and glutamatergic or GABAergic neurotransmission. Expression levels of mRNA of Slc17a6 and Gabrg3, involved in excitation and inhibition respectively, were significantly increased at 28days post-exposure, suggesting a possible role in the CN spontaneous hyperactivity associated with tinnitus and hyperacusis. In the pain and inflammatory array, noise exposure upregulated mRNA expression levels of four pain/inflammatory genes, Tlr2, Oprd1, Kcnq3 and Ntrk1 and decreased mRNA expression levels of two more genes, Ccl12 and Il1β. Pain/inflammatory gene expression changes via Ntrk1 signaling may induce sterile inflammation, neuropathic pain, microglial activation and migration of nerve fibers from the trigeminal, cuneate and vestibular nuclei into the CN. These changes could contribute to somatic tinnitus, hyperacusis and otalgia.
Collapse
Affiliation(s)
- Senthilvelan Manohar
- Center for Hearing & Deafness, State University of New York at Buffalo, Buffalo, NY 14214, United States.
| | - Kimberly Dahar
- Center for Hearing & Deafness, State University of New York at Buffalo, Buffalo, NY 14214, United States
| | - Henry J Adler
- Center for Hearing & Deafness, State University of New York at Buffalo, Buffalo, NY 14214, United States
| | - Ding Dalian
- Center for Hearing & Deafness, State University of New York at Buffalo, Buffalo, NY 14214, United States
| | - Richard Salvi
- Center for Hearing & Deafness, State University of New York at Buffalo, Buffalo, NY 14214, United States
| |
Collapse
|
50
|
Fang L, Fu Y, Zhang TY. Salicylate-Induced Hearing Loss Trigger Structural Synaptic Modifications in the Ventral Cochlear Nucleus of Rats via Medial Olivocochlear (MOC) Feedback Circuit. Neurochem Res 2016; 41:1343-53. [PMID: 26886762 DOI: 10.1007/s11064-016-1836-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 04/13/2015] [Accepted: 04/21/2015] [Indexed: 12/24/2022]
Abstract
Lesion-induced cochlear damage can result in synaptic outgrowth in the ventral cochlear nucleus (VCN). Tinnitus may be associated with the synaptic outgrowth and hyperactivity in the VCN. However, it remains unclear how hearing loss triggers structural synaptic modifications in the VCN of rats subjected to salicylate-induced tinnitus. To address this issue, we evaluated tinnitus-like behavior in rats after salicylate treatment and compared the amplitude of the distortion product evoked otoacoustic emission (DPOAE) and auditory brainstem response (ABR) between control and treated rats. Moreover, we observed the changes in the synaptic ultrastructure and in the expression levels of growth-associated protein (GAP-43), brain-derived neurotrophic factor (BDNF), the microglial marker Iba-1 and glial fibrillary acidic protein (GFAP) in the VCN. After salicylate treatment (300 mg/kg/day for 4 and 8 days), analysis of the gap prepulse inhibition of the acoustic startle showed that the rats were experiencing tinnitus. The changes in the DPOAE and ABR amplitude indicated an improvement in cochlear sensitivity and a reduction in auditory input following salicylate treatment. The treated rats displayed more synaptic vesicles and longer postsynaptic density in the VCN than the control rats. We observed that the GAP-43 expression, predominantly from medial olivocochlear (MOC) neurons, was significantly up-regulated, and that BDNF- and Iba-1-immunoreactive cells were persistently decreased after salicylate administration. Furthermore, GFAP-immunoreactive astrocytes, which is associated with synaptic regrowth, was significantly increased in the treated groups. Our study revealed that reduced auditory nerve activity triggers synaptic outgrowth and hyperactivity in the VCN via a MOC neural feedback circuit. Structural synaptic modifications may be a reflexive process that compensates for the reduced auditory input after salicylate administration. However, massive increases in excitatory synapses in the VCN may represent a detrimental process that causes central hyperactivity, leading to tinnitus.
Collapse
Affiliation(s)
- Lian Fang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Eye and ENT Hospital, Shanghai Medical College, Fudan University, 83 Fenyang Road, Xuhui District, Shanghai, 200031, China
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of WenZhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - YaoYao Fu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Eye and ENT Hospital, Shanghai Medical College, Fudan University, 83 Fenyang Road, Xuhui District, Shanghai, 200031, China
| | - Tian-Yu Zhang
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Eye and ENT Hospital, Shanghai Medical College, Fudan University, 83 Fenyang Road, Xuhui District, Shanghai, 200031, China.
| |
Collapse
|