Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Hear Res ; 449: 109032, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38797035

ABSTRACT

Neurons within a neuronal network can be grouped by bottom-up and top-down influences using synchrony in neuronal oscillations. This creates the representation of perceptual objects from sensory features. Oscillatory activity can be differentiated into stimulus-phase-locked (evoked) and non-phase-locked (induced). The former is mainly determined by sensory input, the latter by higher-level (cortical) processing. Effects of auditory deprivation on cortical oscillations have been studied in congenitally deaf cats (CDCs) using cochlear implant (CI) stimulation. CI-induced alpha, beta, and gamma activity were compromised in the auditory cortex of CDCs. Furthermore, top-down information flow between secondary and primary auditory areas in hearing cats, conveyed by induced alpha oscillations, was lost in CDCs. Here we used the matching pursuit algorithm to assess components of such oscillatory activity in local field potentials recorded in primary field A1. Additionally to the loss of induced alpha oscillations, we also found a loss of evoked theta activity in CDCs. The loss of theta and alpha activity in CDCs can be directly related to reduced high-frequency (gamma-band) activity due to cross-frequency coupling. Here we quantified such cross-frequency coupling in adult 1) hearing-experienced, acoustically stimulated cats (aHCs), 2) hearing-experienced cats following acute pharmacological deafening and subsequent CIs, thus in electrically stimulated cats (eHCs), and 3) electrically stimulated CDCs. We found significant cross-frequency coupling in all animal groups in > 70% of auditory-responsive sites. The predominant coupling in aHCs and eHCs was between theta/alpha phase and gamma power. In CDCs such coupling was lost and replaced by alpha oscillations coupling to delta/theta phase. Thus, alpha/theta oscillations synchronize high-frequency gamma activity only in hearing-experienced cats. The absence of induced alpha and theta oscillations contributes to the loss of induced gamma power in CDCs, thereby signifying impaired local network activity.


Subject(s)
Acoustic Stimulation , Auditory Cortex , Deafness , Gamma Rhythm , Animals , Cats , Auditory Cortex/physiopathology , Deafness/physiopathology , Deafness/congenital , Cochlear Implants , Alpha Rhythm , Evoked Potentials, Auditory , Algorithms , Auditory Pathways/physiopathology , Disease Models, Animal , Theta Rhythm
2.
MethodsX ; 12: 102674, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38660047

ABSTRACT

The neocortex of the brain can be divided into six layers each with a distinct cell composition and connectivity pattern. Recently, sensory deprivation, including congenital deafness, has been shown to alter cortical structure (e.g. the cortical thickness) of the feline auditory cortex with variable and inconsistent results. Thus, understanding these complex changes will require further study of the constituent cortical layers in three-dimensional space. Further progress crucially depends on the use of objective computational techniques that can reliably characterize spatial properties of the complex cortical structure. Here a method for cortical laminar segmentation is derived and applied to the three-dimensional cortical areas reconstructed from a series of histological sections from four feline brains. In this approach, the Alternating Kernel Method was extended to fit a multi-variate Gaussian mixture model to a feature space consisting of both staining intensity and a biologically plausible equivolumetric depth map. This research method•Extends the Alternating Kernel Method to multi-dimensional feature spaces.•Uses it to segment the cortical layers in reconstructed histology volume. Segmentation features include staining intensity and a biologically plausible equivolumetric depth map.•Validates results in auditory cortical areas of feline brains, two with normal hearing and two with congenital deafness.

3.
Curr Res Neurobiol ; 6: 100124, 2024.
Article in English | MEDLINE | ID: mdl-38616957

ABSTRACT

Background: In children, hearing loss has been associated with hyperactivity, disturbed social interaction, and risk of cognitive disturbances. Mechanistic explanations of these relations sometimes involve language. To investigate the effect of hearing loss on behavioral deficits in the absence of language, we tested the impact of hearing loss in juvenile rats on motor, social, and cognitive behavior and on physiology of prefrontal cortex. Methods: Hearing loss was induced in juvenile (postnatal day 14) male Sprague-Dawley rats by intracochlear injection of neomycin under general anesthesia. Sham-operated and non-operated hearing rats served as controls. One week after surgery auditory brainstem response (ABR) measurements verified hearing loss or intact hearing in sham-operated and non-operated controls. All rats were then tested for locomotor activity (open field), coordination (Rotarod), and for social interaction during development in weeks 1, 2, 4, 8, 16, and 24 after surgery. From week 8 on, rats were trained and tested for spatial learning and memory (4-arm baited 8-arm radial maze test). In a final setting, neuronal activity was recorded in the medial prefrontal cortex (mPFC). Results: In the open field deafened rats moved faster and covered more distance than sham-operated and non-operated controls from week 8 on (both p < 0.05). Deafened rats showed significantly more play fighting during development (p < 0.05), whereas other aspects of social interaction, such as following, were not affected. Learning of the radial maze test was not impaired in deafened rats (p > 0.05), but rats used less next-arm entries than other groups indicating impaired concept learning (p < 0.05). In the mPFC neuronal firing rate was reduced and enhanced irregular firing was observed. Moreover, oscillatory activity was altered, both within the mPFC and in coherence of mPFC with the somatosensory cortex (p < 0.05). Conclusions: Hearing loss in juvenile rats leads to hyperactive behavior and pronounced play-fighting during development, suggesting a causal relationship between hearing loss and cognitive development. Altered neuronal activities in the mPFC after hearing loss support such effects on neuronal networks outside the central auditory system. This animal model provides evidence of developmental consequences of juvenile hearing loss on prefrontal cortex in absence of language as potential confounding factor.

4.
Hear Res ; 443: 108953, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38277881

ABSTRACT

The present study investigates effects of current focusing and pulse shape on threshold, dynamic range, spread of excitation and channel interaction in the time domain using cochlear implant stimulation. The study was performed on 20 adult guinea pigs using a 6-channel animal cochlear implant, recording was performed in the auditory midbrain using a multielectrode array. After determining the best frequencies for individual recording contacts with acoustic stimulation, the ear was deafened and a cochlear implant was inserted into the cochlea. The position of the implant was controlled by x-ray. Stimulation with biphasic, pseudomonophasic and monophasic stimuli was performed with monopolar, monopolar with common ground, bipolar and tripolar configuration in two sets of experiments, allowing comparison of the effects of the different stimulation strategies on threshold, dynamic range, spread of excitation and channel interaction. Channel interaction was studied in the temporal domain, where two electrodes were activated with pulse trains and phase locking to these pulse trains in the midbrain was quantified. The results documented multifactorial influences on the response properties, with significant interaction between factors. Thresholds increased with increasing current focusing, but decreased with pseudomonophasic and monophasic pulse shapes. The results documented that current focusing, particularly tripolar configuration, effectively reduces channel interaction, but that also pseudomonophasic and monophasic stimulation and phase duration intensity coding reduce channel interactions.


Subject(s)
Cochlear Implantation , Cochlear Implants , Deafness , Animals , Guinea Pigs , Auditory Threshold/physiology , Deafness/rehabilitation , Cochlea/physiology , Electric Stimulation
5.
J Speech Lang Hear Res ; 67(2): 618-632, 2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38198368

ABSTRACT

OBJECTIVES: The primary aim was to investigate the variability in language development in children aged 5-7.5 years after bilateral cochlear implantation (CI) up to the age of 2 years, and any impact of the age at implantation and additional noncognitive or anatomical disorders at implantation. DESIGN: Data of 84 congenitally deaf children that had received simultaneous bilateral CI at the age of ≤ 24 months were included in this retrospective study. The results of language comprehension acquisition were evaluated using a standardized German language acquisition test for normal hearing preschoolers and first graders. Data on speech perception of monosyllables and sentences in quiet and noise were added. RESULTS: In a monosyllabic test, the children achieved a median performance of 75.0 ± 12.88%. In the sentence test in quiet, the median performance was 89 ± 12.69%, but dropped to 54 ± 18.92% in noise. A simple analysis showed a significant main effect of age at implantation on monosyllabic word comprehension (p < .001), but no significant effect of comorbidities that lacked cognitive effects (p = .24). Language acquisition values correspond to the normal range of children with normal hearing. Approximately 25% of the variability in the language acquisition tests is due to the outcome of the monosyllabic speech perception test. CONCLUSIONS: Congenitally deaf children who were fitted bilaterally in the 1st year of life can develop age-appropriate language skills by the time they start school. The high variability in the data is partly due to the age of implantation, but additional factors such as cognitive factors (e.g., working memory) are likely to influence the variability.


Subject(s)
Cochlear Implantation , Cochlear Implants , Deafness , Speech Perception , Child , Humans , Cochlear Implantation/methods , Retrospective Studies , Language Development , Deafness/surgery , Treatment Outcome
6.
Sci Rep ; 13(1): 22657, 2023 12 19.
Article in English | MEDLINE | ID: mdl-38114599

ABSTRACT

Vibrotactile stimulation is believed to enhance auditory speech perception, offering potential benefits for cochlear implant (CI) users who may utilize compensatory sensory strategies. Our study advances previous research by directly comparing tactile speech intelligibility enhancements in normal-hearing (NH) and CI participants, using the same paradigm. Moreover, we assessed tactile enhancement considering stimulus non-specific, excitatory effects through an incongruent audio-tactile control condition that did not contain any speech-relevant information. In addition to this incongruent audio-tactile condition, we presented sentences in an auditory only and a congruent audio-tactile condition, with the congruent tactile stimulus providing low-frequency envelope information via a vibrating probe on the index fingertip. The study involved 23 NH listeners and 14 CI users. In both groups, significant tactile enhancements were observed for congruent tactile stimuli (5.3% for NH and 5.4% for CI participants), but not for incongruent tactile stimulation. These findings replicate previously observed tactile enhancement effects. Juxtaposing our study with previous research, the informational content of the tactile stimulus emerges as a modulator of intelligibility: Generally, congruent stimuli enhanced, non-matching tactile stimuli reduced, and neutral stimuli did not change test outcomes. We conclude that the temporal cues provided by congruent vibrotactile stimuli may aid in parsing continuous speech signals into syllables and words, consequently leading to the observed improvements in intelligibility.


Subject(s)
Cochlear Implantation , Cochlear Implants , Speech Perception , Humans , Speech Intelligibility , Acoustic Stimulation , Auditory Perception/physiology , Speech Perception/physiology
7.
Sci Rep ; 13(1): 15787, 2023 09 22.
Article in English | MEDLINE | ID: mdl-37737452

ABSTRACT

Environmental events often occur on a probabilistic basis but can sometimes be predicted based on specific cues and thus approached proactively. Incidental statistical learning enables the acquisition of knowledge about probabilistic cue-target contingencies. However, the neural mechanisms of statistical learning about contingencies (SLC), the required conditions for successful learning, and the role of implicit processes in the resultant proactive behavior are still debated. We examined changes in behavior and cortical activity during an SLC task in which subjects responded to visual targets. Unbeknown to them, there were three types of target cues associated with high-, low-, and zero target probabilities. About half of the subjects spontaneously gained explicit knowledge about the contingencies (contingency-aware group), and only they showed evidence of proactivity: shortened response times to predictable targets and enhanced event-related brain responses (cue-evoked P300 and contingent negative variation, CNV) to high probability cues. The behavioral and brain responses were strictly associated on a single-trial basis. Source reconstruction of the brain responses revealed activation of fronto-parietal brain regions associated with cognitive control, particularly the anterior cingulate cortex and precuneus. We also found neural correlates of SLC in the contingency-unaware group, but these were restricted to post-target latencies and visual association areas. Our results document a qualitative difference between explicit and implicit learning processes and suggest that in certain conditions, proactivity may require explicit knowledge about contingencies.


Subject(s)
Brain , Learning , Humans , Cues , Awareness , Electroencephalography
8.
Laryngorhinootologie ; 102(S 01): S3-S11, 2023 05.
Article in English, German | MEDLINE | ID: mdl-37130527

ABSTRACT

The human brain shows extensive development of the cerebral cortex after birth. This is extensively altered by the absence of auditory input: the development of cortical synapses in the auditory system is delayed and their degradation is increased. Recent work shows that the synapses responsible for corticocortical processing of stimuli and their embedding into multisensory interactions and cognition are particularly affected. Since the brain is heavily reciprocally interconnected, inborn deafness manifests not only in deficits in auditory processing, but also in cognitive (non-auditory) functions that are affected differently between individuals. It requires individualized approaches in therapy of deafness in childhood.


Subject(s)
Auditory Cortex , Deafness , Humans , Auditory Perception , Cognition , Deafness/psychology , Hearing
9.
Hear Res ; 433: 108763, 2023 06.
Article in English | MEDLINE | ID: mdl-37104991

ABSTRACT

The extent to which aging of the central auditory pathway impairs auditory perception in the elderly independent of peripheral cochlear decline is debated. To cause auditory deficits in normal hearing elderly, central aging needs to degrade neural sound representations at some point along the auditory pathway. However, inaccessible to psychophysical methods, the level of the auditory pathway at which aging starts to effectively degrade neural sound representations remains poorly differentiated. Here we tested how potential age-related changes in the auditory brainstem affect the stability of spatiotemporal multiunit complex speech-like sound representations in the auditory midbrain of old normal hearing CBA/J mice. Although brainstem conduction speed slowed down in old mice, the change was limited to the sub-millisecond range and only minimally affected temporal processing in the midbrain (i.e. gaps-in-noise sensitivity). Importantly, besides the small delay, multiunit complex temporal sound representations in the auditory midbrain did not differ between young and old mice. This shows that although small age-related neural effects in simple sound parameters in the lower brainstem may be present in aging they do not effectively deteriorate complex neural population representations at the level of the auditory midbrain when peripheral hearing remains normal. This result challenges the widespread belief of 'pure' central auditory decline as an automatic consequence of aging, at least up to the inferior colliculus. However, the stability of midbrain processing in aging emphasizes the role of undetected 'hidden' peripheral damage and accumulating effects in higher cortical auditory-cognitive processing explaining perception deficits in 'normal hearing' elderly.


Subject(s)
Longevity , Mesencephalon , Mice , Animals , Mice, Inbred CBA , Auditory Perception , Brain Stem , Aging/psychology , Evoked Potentials, Auditory, Brain Stem
10.
Trends Neurosci ; 46(5): 377-393, 2023 05.
Article in English | MEDLINE | ID: mdl-36990952

ABSTRACT

Crossmodal plasticity is a textbook example of the ability of the brain to reorganize based on use. We review evidence from the auditory system showing that such reorganization has significant limits, is dependent on pre-existing circuitry and top-down interactions, and that extensive reorganization is often absent. We argue that the evidence does not support the hypothesis that crossmodal reorganization is responsible for closing critical periods in deafness, and crossmodal plasticity instead represents a neuronal process that is dynamically adaptable. We evaluate the evidence for crossmodal changes in both developmental and adult-onset deafness, which start as early as mild-moderate hearing loss and show reversibility when hearing is restored. Finally, crossmodal plasticity does not appear to affect the neuronal preconditions for successful hearing restoration. Given its dynamic and versatile nature, we describe how this plasticity can be exploited for improving clinical outcomes after neurosensory restoration.


Subject(s)
Auditory Cortex , Deafness , Hearing Loss , Adult , Humans , Auditory Cortex/physiology , Acoustic Stimulation , Photic Stimulation , Neuronal Plasticity/physiology
11.
Hear Res ; 429: 108687, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36638762

ABSTRACT

In cochlear implantation, current preoperative planning procedures allow for estimating how far a specific implant will reach into the inner ear of the patient, which is important to optimize hearing preservation and speech perception outcomes. Here we report on the development of a methodology that goes beyond current planning approaches: the proposed model does not only estimate specific outcome parameters but allows for entire, three-dimensional virtual implantations of patient-specific cochlear anatomies with different types of electrode arrays. The model was trained based on imaging datasets of 186 human cochleae, which contained 171 clinical computer tomographies (CTs) of actual cochlear implant patients as well as 15 high-resolution micro-CTs of cadaver cochleae to also reconstruct the refined intracochlear structures not visible in clinical imaging. The model was validated on an independent dataset of 141 preoperative and postoperative clinical CTs of cochlear implant recipients and outperformed all currently available planning approaches, not only in terms of accuracy but also regarding the amount of information that is available prior to the actual implantation.


Subject(s)
Cochlear Implantation , Cochlear Implants , Hearing Loss, Sensorineural , Speech Perception , Humans , Cochlear Implantation/methods , Hearing Loss, Sensorineural/rehabilitation , Cochlea/diagnostic imaging , Cochlea/surgery , Hearing
12.
Ear Hear ; 44(1): 118-134, 2023.
Article in English | MEDLINE | ID: mdl-35894668

ABSTRACT

OBJECTIVES: Cochlear implantation criteria include subjects with residual low-frequency hearing. To minimize implantation trauma and to avoid unwanted interactions of electric- and acoustic stimuli, it is often recommended to stop cochlear implantation before the cochlear implant (CI) reaches the cochlear partition with residual hearing, as determined by an audiogram. For this purpose, the implant can be used to record acoustically evoked signals during implantation, including cochlear compound action potentials (CAP), cochlear microphonics (CMs), and summating potentials (SPs). The former two have previously been used to monitor residual hearing in clinical settings. DESIGN: In the present study we investigated the use of intracochlear, bipolar SP recordings to determine the exact cochlear position of the contacts of implanted CIs in guinea pig cochleae (n = 13). Polarity reversals of SPs were used as a functional marker of intracochlear position. Micro computed tomography (µCT) imaging and a modified Greenwood function were used to determine the cochleotopic positions of the contacts in the cochlea. These anatomical reconstructions were used to validate the SP-based position estimates. RESULTS: The precision of the SP-based position estimation was on average within ± 0.37 octaves and was not impaired by moderate hearing loss caused by noise exposure after implantation. It is important to note that acute hearing impairment did not reduce the precision of the method. The cochleotopic position of CI accounted for ~70% of the variability of SP polarity reversals. Outliers in the dataset were associated with lateral CI positions. Last, we propose a simplified method to avoid implantation in functioning parts of the cochlea by approaching a predefined frequency region using bipolar SP recordings through a CI. CONCLUSIONS: Bipolar SP recordings provide reliable information on electrode position in the cochlea. The position estimate remains reliable after moderate hearing loss. The technique presented here could be applied during CI surgery to monitor the CI approach to a predefined frequency region.


Subject(s)
Cochlear Implantation , Cochlear Implants , Deafness , Hearing Loss, Sudden , Animals , Guinea Pigs , Audiometry, Evoked Response/methods , X-Ray Microtomography , Cochlear Implantation/methods , Cochlea , Deafness/rehabilitation
13.
J Assoc Res Otolaryngol ; 24(1): 31-46, 2023 02.
Article in English | MEDLINE | ID: mdl-36459250

ABSTRACT

One severe side effect of the use of cochlear implants (CI) is coincidental facial nerve stimulation (FNS). Clinical methods to alleviate FNS range from the reprogramming of processor settings to revision surgery. We systematically assessed different changes in CI stimulation modes that have been discussed in the literature as "rescue factors" from FNS: electrode configuration (broad to focused), pulse shape (symmetric biphasic to pseudo-monophasic), and pulse polarity (cathodic to anodic). An FNS was assessed, based on electrophysiological thresholds, in 204 electrically evoked compound action potential (eCAP) input/output functions recorded from 33 ears of 26 guinea pigs. The stimulation level difference between auditory nerve eCAP threshold and FNS threshold was expressed as the eCAP-to-FNS offset. Coincidental FNS occurred in all animals and in 45% of all recordings. A change from monopolar to focused (bipolar, tripolar) configurations minimized FNS. The Euclidean distance between the CI contacts and the facial nerve explained no more than 33% of the variance in FNS thresholds. For both the FNS threshold and the eCAP-to-FNS offset, the change from cathodic to anodic pulse polarity significantly reduced FNS and permitted a gain of 14-71% of the dynamic range of the eCAP response. This "anodic rescue effect" was stronger for pseudo-monophasic pulses as compared to the symmetric biphasic pulse shape. These results provide possible mechanisms underlying recent clinical interventions to alleviate FNS. The "anodic-rescue effect" may offer a non-invasive therapeutic option for FNS in human CI users that should be tested clinically, preferably in combination with current-focusing methods.


Subject(s)
Cochlear Implantation , Cochlear Implants , Humans , Animals , Guinea Pigs , Facial Nerve , Cochlear Implants/adverse effects , Cochlea/physiology , Evoked Potentials , Action Potentials/physiology , Electric Stimulation/methods , Cochlear Nerve , Evoked Potentials, Auditory/physiology
14.
Front Neurosci ; 16: 966568, 2022.
Article in English | MEDLINE | ID: mdl-36440269

ABSTRACT

Background: Hearing loss was proposed as a factor affecting development of cognitive impairment in elderly. Deficits cannot be explained primarily by dysfunctional neuronal networks within the central auditory system. We here tested the impact of hearing loss in adult rats on motor, social, and cognitive function. Furthermore, potential changes in the neuronal activity in the medial prefrontal cortex (mPFC) and the inferior colliculus (IC) were evaluated. Materials and methods: In adult male Sprague Dawley rats hearing loss was induced under general anesthesia with intracochlear injection of neomycin. Sham-operated and naive rats served as controls. Postsurgical acoustically evoked auditory brainstem response (ABR)-measurements verified hearing loss after intracochlear neomycin-injection, respectively, intact hearing in sham-operated and naive controls. In intervals of 8 weeks and up to 12 months after surgery rats were tested for locomotor activity (open field) and coordination (Rotarod), for social interaction and preference, and for learning and memory (4-arms baited 8-arms radial maze test). In a final setting, electrophysiological recordings were performed in the mPFC and the IC. Results: Locomotor activity did not differ between deaf and control rats, whereas motor coordination on the Rotarod was disturbed in deaf rats (P < 0.05). Learning the concept of the radial maze test was initially disturbed in deaf rats (P < 0.05), whereas retesting every 8 weeks did not show long-term memory deficits. Social interaction and preference was also not affected by hearing loss. Final electrophysiological recordings in anesthetized rats revealed reduced firing rates, enhanced irregular firing, and reduced oscillatory theta band activity (4-8 Hz) in the mPFC of deaf rats as compared to controls (P < 0.05). In the IC, reduced oscillatory theta (4-8 Hz) and gamma (30-100 Hz) band activity was found in deaf rats (P < 0.05). Conclusion: Minor and transient behavioral deficits do not confirm direct impact of long-term hearing loss on cognitive function in rats. However, the altered neuronal activities in the mPFC and IC after hearing loss indicate effects on neuronal networks in and outside the central auditory system with potential consequences on cognitive function.

15.
Braz. j. otorhinolaryngol. (Impr.) ; 88(4): 546-555, July-Aug. 2022. graf
Article in English | LILACS-Express | LILACS | ID: biblio-1394148

ABSTRACT

Abstract Introduction: Electrocochleography has recently emerged as a diagnostic tool in cochlear implant surgery, purposing hearing preservation and optimal electrode positioning. Objective: In this experimental study, extra-cochlear potentials were obtained during cochlear implant surgery in guinea pigs. The aim was to determine electrophysiological changes indicating cochlear trauma after cochleostomy and after electrode implantation in different insertion depths. Methods: Normal-hearing guinea pigs (n = 14) were implanted uni- or bilaterally with a multichannel electrode. The extra-cochlear cochlear nerve action potentials were obtained in response to acoustic stimuli at specific frequencies before and after cochleostomy, and after introduction of the electrode bundle. After the electrophysiological experiments, the guinea pigs were euthanized and microtomography was performed, in order to determine the position of the electrode and to calculate of the depth of insertion. Based on the changes of amplitude and thresholds in relation to the stimulus frequency, the electrophysiological data and the position obtained by the microtomography reconstruction were compared. Results: Cochleostomy promoted a small electrophysiological impact, while electrode insertion caused changes in the amplitude of extra-cochlear electrophysiological potentials over a wide range of frequencies, especially in the deepest insertions. There was, however, preservation of the electrical response to low frequency stimuli in most cases, indicating a limited auditory impact in the intraoperative evaluation. The mean insertion depth of the apical electrodes was 5339.56 μm (±306.45 - 6 inserted contacts) and 4447.75 μm (±290.23 - 5 inserted contacts). Conclusions: The main electrophysiological changes observed during surgical procedures occurred during implantation of the electrode, especially the deepest insertions, whereas the cochleostomy disturbed the potentials to a lesser extent. While hearing loss was often observed apical to the cochlear implant, it was possible to preserve low frequencies after insertion. © 2020 Associação Brasileira de Otorrinolaringologia e Cirurgia Cérvico-Facial. Published by Elsevier Editora Ltda. This is an open access article under the CC BY license (http:// creativecommons.org/licenses/by/4.0/).


Resumo Introdução: A eletrococleografia surgiu recentemente como uma ferramenta diagnóstica na cirurgia de implante coclear, objetiva a preservação da audição e o posicionamento ideal dos eletrodos. Objetivo: Determinar as alterações eletrofisiológicas indicativas de trauma coclear após a cocleostomia e após o implante do eletrodo em diferentes profundidades de inserção. Método: Neste estudo experimental, potenciais extracocleares foram obtidos durante a cirurgia de implante coclear em cobaias. Cobaias com audição normal (n = 14) foram implantadas uni- ou bilateralmente com eletrodo multicanal. Os potenciais de ação do nervo coclear extracoclear foram obtidos em resposta a estímulos acústicos em frequências específicas antes e após a cocleostomia e após a introdução do feixe de eletrodos. Após os experimentos eletrofisiológicos, as cobaias foram submetidas à eutanásia e a microtomografia foi feita para determinar a posição do eletrodo e calcular a profundidade de inserção. Com base nas mudanças de amplitude e limiares em relação à frequência do estímulo, os dados eletrofisiológicos e a posição obtida na reconstrução microtomográfica foram comparados. Resultados: A cocleostomia promoveu um pequeno impacto eletrofisiológico, enquanto a inserção do eletrodo causou alterações na amplitude dos potenciais eletrofisiológicos extra-cocleares em uma ampla faixa de frequências, especialmente nas inserções mais profundas. Houve, entretanto, preservação da resposta elétrica aos estímulos de baixa frequência na maioria dos casos, indicou um impacto auditivo limitado na avaliação intraoperatória. A profundidade média de inserção dos eletrodos apicais foi 5339,56 μm (± 306,45 - 6 contatos inseridos) e 4447,75 μm (± 290,23 - 5 contatos inseridos). Conclusão: As principais alterações eletrofisiológicas observadas durante os procedimentos cirúrgicos ocorreram durante o implante do eletrodo, especialmente nas inserções mais profundas, enquanto a cocleostomia alterou os potenciais em menor grau. Embora a perda auditiva seja frequentemente observada em posição apical ao implante coclear, foi possível preservar as baixas frequências após a inserção.

16.
J Vis Exp ; (184)2022 06 23.
Article in English | MEDLINE | ID: mdl-35815969

ABSTRACT

Visual evoked potentials (VEP) allow the characterization of visual function in preclinical mouse models. Various methods exist to measure VEPs in mice, from non-invasive EEG, subcutaneous single-electrodes, and ECoG to fully invasive intracortical multichannel visual cortex recordings. It can be useful to acquire a global, topographical EEG-level characterization of visual responses previous to local intracortical microelectrode measurements in acute experimental settings. For example, one use case is to assess global cross-modal changes in VEP topography in deafness models before studying its effects on a local intracortical level. Multichannel epicranial EEG is a robust method to acquire such an overview measure of cortical visual activity. Multichannel epicranial EEG provides comparable results through a standardized, consistent approach to, for example, identify cross-modal, pathological, or age-related changes in cortical visual function. The current study presents a method to obtain the topographical distribution of flash-evoked VEPs with a 32-channel thin-film EEG electrode array in anesthetized mice. Combined with analysis in the time and frequency domain, this approach allows fast characterization and screening of the topography and basic visual properties of mouse cortical visual function, which can be combined with various acute experimental settings.


Subject(s)
Evoked Potentials, Visual , Visual Cortex , Animals , Electrodes , Electroencephalography/methods , Mice , Neurologic Examination , Photic Stimulation/methods , Visual Cortex/physiology
17.
Sci Rep ; 12(1): 5047, 2022 03 23.
Article in English | MEDLINE | ID: mdl-35322066

ABSTRACT

Cochlear variability is of key importance for the clinical use of cochlear implants, the most successful neuroprosthetic device that is surgically placed into the cochlear scala tympani. Despite extensive literature on human cochlear variability, few information is available on the variability of the modiolar wall. In the present study, we analyzed 108 corrosion casts, 95 clinical cone beam computer tomographies (CTs) and 15 µCTs of human cochleae and observed modiolar variability of similar and larger extent than the lateral wall variability. Lateral wall measures correlated with modiolar wall measures significantly. ~ 49% of the variability had a common cause. Based on these data we developed a model of the modiolar wall variations and related the model to the design of cochlear implants aimed for perimodiolar locations. The data demonstrate that both the insertion limits relevant for lateral wall damage (approximate range of 4-9 mm) as well as the dimensions required for optimal perimodiolar placement of the electrode (the point of release from the straightener; approximate range of 2-5mm) are highly interindividually variable. The data demonstrate that tip fold-overs of preformed implants likely result from the morphology of the modiolus (with radius changing from base to apex), and that optimal cochlear implantation of perimodiolar arrays cannot be guaranteed without an individualized surgical technique.


Subject(s)
Cochlear Implantation , Cochlear Implants , Cochlea/anatomy & histology , Cochlea/diagnostic imaging , Cochlea/surgery , Cochlear Implantation/methods , Cone-Beam Computed Tomography , Electrodes, Implanted , Humans , Scala Tympani/surgery
18.
Front Syst Neurosci ; 16: 806142, 2022.
Article in English | MEDLINE | ID: mdl-35283734

ABSTRACT

The influence of sensory experience on cortical feedforward and feedback interactions has rarely been studied in the auditory cortex. Previous work has documented a dystrophic effect of deafness in deep cortical layers, and a reduction of interareal couplings between primary and secondary auditory areas in congenital deafness which was particularly pronounced in the top-down direction (from the secondary to the primary area). In the present study, we directly quantified the functional interaction between superficial (supragranular, I to III) and deep (infragranular, V and VI) layers of feline's primary auditory cortex A1, and also between superficial/deep layers of A1 and a secondary auditory cortex, namely the posterior auditory field (PAF). We compared adult hearing cats under acoustic stimulation and cochlear implant (CI) stimulation to adult congenitally deaf cats (CDC) under CI stimulation. Neuronal activity was recorded from auditory fields A1 and PAF simultaneously with two NeuroNexus electrode arrays. We quantified the spike field coherence (i.e., the statistical dependence of spike trains at one electrode with local field potentials on another electrode) using pairwise phase consistency (PPC). Both the magnitude as well as the preferred phase of synchronization was analyzed. The magnitude of PPC was significantly smaller in CDCs than in controls. Furthermore, controls showed no significant difference between the preferred phase of synchronization between supragranular and infragranular layers, both in acoustic and electric stimulation. In CDCs, however, there was a large difference in the preferred phase between supragranular and infragranular layers. These results demonstrate a loss of synchrony and for the first time directly document a functional decoupling of the interaction between supragranular and infragranular layers of the primary auditory cortex in congenital deafness. Since these are key for the influence of top-down to bottom-up computations, the results suggest a loss of recurrent cortical processing in congenital deafness and explain the outcomes of previous studies by deficits in intracolumnar microcircuitry.

19.
Neuroimage Clin ; 33: 102942, 2022.
Article in English | MEDLINE | ID: mdl-35033811

ABSTRACT

In naturalistic situations, sounds are often perceived in conjunction with matching visual impressions. For example, we see and hear the neighbor's dog barking in the garden. Still, there is a good chance that we recognize the neighbor's dog even when we only hear it barking, but do not see it behind the fence. Previous studies with normal-hearing (NH) listeners have shown that the audio-visual presentation of a perceptual object (like an animal) increases the probability to recognize this object later on, even if the repeated presentation of this object occurs in a purely auditory condition. In patients with a cochlear implant (CI), however, the electrical hearing of sounds is impoverished, and the ability to recognize perceptual objects in auditory conditions is significantly limited. It is currently not well understood whether CI users - as NH listeners - show a multisensory facilitation for auditory recognition. The present study used event-related potentials (ERPs) and a continuous recognition paradigm with auditory and audio-visual stimuli to test the prediction that CI users show a benefit from audio-visual perception. Indeed, the congruent audio-visual context resulted in an improved recognition ability of objects in an auditory-only condition, both in the NH listeners and the CI users. The ERPs revealed a group-specific pattern of voltage topographies and correlations between these ERP maps and the auditory recognition ability, indicating a different processing of congruent audio-visual stimuli in CI users when compared to NH listeners. Taken together, our results point to distinct cortical processing of naturalistic audio-visual objects in CI users and NH listeners, which however allows both groups to improve the recognition ability of these objects in a purely auditory context. Our findings are of relevance for future clinical research since audio-visual perception might also improve the auditory rehabilitation after cochlear implantation.


Subject(s)
Cochlear Implantation , Cochlear Implants , Speech Perception , Acoustic Stimulation , Aged , Auditory Perception , Evoked Potentials , Humans , Recognition, Psychology , Visual Perception
20.
Neurobiol Aging ; 110: 47-60, 2022 02.
Article in English | MEDLINE | ID: mdl-34852306

ABSTRACT

Impaired temporal resolution of the central auditory system has long been suggested to contribute to speech understanding deficits in the elderly. However, it has been difficult to differentiate between direct age-related central deficits and indirect effects of confounding peripheral age-related hearing loss on temporal resolution. To differentiate this, we measured temporal acuity in the inferior colliculus (IC) of aged CBA/J and C57BL/6 mice, as a model of aging with and without concomitant hearing loss. We used two common measures of auditory temporal processing: gap detection as a measure of temporal fine structure and amplitude-modulated noise as a measure of envelope sensitivity. Importantly, auditory temporal acuity remained precise in the IC of old CBA/J mice when no or only minimal age-related hearing loss was present. In contrast, temporal acuity was only indirectly reduced by the presence of age-related hearing loss in aged C57BL/6 mice, not by affecting the brainstem precision, but by affecting the signal-to-noise ratio of the neuronal activity in the IC. This demonstrates that indirect effects of age-related peripheral hearing loss likely remain an important factor for temporal processing in aging in comparison to 'pure' central auditory decline itself. It also draws attention to the issue that the threshold difference between 'nearly normal' or 'clinically normal' hearing aging subjects in comparison to normal hearing young subjects still can have indirect effects on central auditory neural representations of temporal processing.


Subject(s)
Aging/physiology , Auditory Perception/physiology , Inferior Colliculi/physiology , Presbycusis/physiopathology , Time Perception/physiology , Animals , Mice, Inbred C57BL , Mice, Inbred CBA , Neurons/physiology , Presbycusis/etiology , Signal-To-Noise Ratio
SELECTION OF CITATIONS
SEARCH DETAIL
...