Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Hear Res ; 439: 108895, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37837701

RESUMO

The auditory nerve typically degenerates following loss of cochlear hair cells or synapses. In the case of hair cell loss neural degeneration hinders restoration of hearing through a cochlear implant, and in the case of synaptopathy suprathreshold hearing is affected, potentially degrading speech perception in noise. It has been established that neurotrophins such as brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3) can mitigate auditory nerve degeneration. Several potential BDNF mimetics have also been investigated for neurotrophic effects in the cochlea. A recent in vitro study showed favorable effects of M3, a TrkB monoclonal antibody agonist, when compared with BDNF. In the present study we set out to examine the effect of M3 on auditory nerve preservation in vivo. Thirty-one guinea pigs were bilaterally deafened, and unilaterally treated with a single 3-µl dose of 7 mg/ml, 0.7 mg/ml M3 or vehicle-only by means of a small gelatin sponge two weeks later. During the experiment and analyses the experimenters were blinded to the three treatment groups. Four weeks after treatment, we assessed the treatment effect (1) histologically, by quantifying survival of SGCs and their peripheral processes (PPs); and (2) electrophysiologically, with two different paradigms of electrically evoked compound action potential (eCAP) recordings shown to be indicative of neural health: single-pulse stimulation with varying inter-phase gap (IPG), and pulse-train stimulation with varying inter-pulse interval. We observed a consistent and significant preservative effect of M3 on SGC survival in the lower basal turn (approximately 40% more survival than in the untreated contralateral cochlea), but also in the upper middle and lower apical turn of the cochlea. This effect was similar for the two treatment groups. Survival of PPs showed a trend similar to that of the SGCs, but was only significantly higher for the highest dose of M3. The protective effect of M3 on SGCs was not reflected in any of the eCAP measures: no statistically significant differences were observed between groups in IPG effect nor between the M3 treatment groups and the control group using the pulse-train stimulation paradigm. In short, while a clear effect of M3 was observed on SGC survival, this was not clearly translated into functional preservation.


Assuntos
Implantes Cocleares , Surdez , Cobaias , Animais , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Gânglio Espiral da Cóclea/patologia , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Nervo Coclear , Audição , Cóclea
2.
Front Cell Neurosci ; 17: 1236894, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37692553

RESUMO

Introduction: The leucine-rich repeat-containing G-protein coupled receptor 5 (LGR5) is a tissue resident stem cell marker, which it is expressed in supporting cells (SCs) in the organ of Corti in the mammalian inner ear. These LGR5+ SCs can be used as an endogenous source of progenitor cells for regeneration of hair cells (HCs) to treat hearing loss and deafness. We have recently reported that LGR5+ SCs survive 1 week after ototoxic trauma. Here, we evaluated Lgr5 expression in the adult cochlea and long-term survival of LGR5+ SCs following severe hearing loss. Methods: Lgr5GFP transgenic mice and wild type mice aged postnatal day 30 (P30) and P200 were used. P30 animals were deafened with a single dose of furosemide and kanamycin. Seven and 28 days after deafening, auditory brainstem responses (ABRs) were recorded. Cochleas were harvested to characterize mature HCs and LGR5+ SCs by immunofluorescence microscopy and quantitative reverse transcription PCR (q-RT-PCR). Results: There were no significant age-related changes in Lgr5 expression when comparing normal-hearing (NH) mice aged P200 with P30. Seven and 28 days after ototoxic trauma, there was severe outer HC loss and LGR5 was expressed in the third row of Deiters' cells and in inner pillar cells. Seven days after induction of ototoxic trauma there was an up-regulation of the mRNA expression of Lgr5 compared to the NH condition; 28 days after ototoxic trauma Lgr5 expression was similar to NH levels. Discussion: The presence of LGR5+ SCs in the adult mouse cochlea, which persists after severe HC loss, suggests potential regenerative capacity of endogenous cochlear progenitor cells in adulthood. To our knowledge, this is the first study showing not only long-term survival of LGR5+ SCs in the normal and ototoxically damaged cochlea, but also increased Lgr5 expression in the adult mouse cochlea after deafening, suggesting long-term availability of potential target cells for future regenerative therapies.

3.
Front Neurosci ; 17: 978230, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36845413

RESUMO

Introduction: Electrocochleography (ECochG) is increasingly used in cochlear implant (CI) surgery, in order to monitor the effect of insertion of the electrode array aiming to preserve residual hearing. However, obtained results are often difficult to interpret. Here we aim to relate changes in ECochG responses to acute trauma induced by different stages of cochlear implantation by performing ECochG at multiple time points during the procedure in normal-hearing guinea pigs. Materials and methods: Eleven normal-hearing guinea pigs received a gold-ball electrode that was fixed in the round-window niche. ECochG recordings were performed during the four steps of cochlear implantation using the gold-ball electrode: (1) Bullostomy to expose the round window, (2) hand-drilling of 0.5-0.6 mm cochleostomy in the basal turn near the round window, (3) insertion of a short flexible electrode array, and (4) withdrawal of electrode array. Acoustical stimuli were tones varying in frequency (0.25-16 kHz) and sound level. The ECochG signal was primarily analyzed in terms of threshold, amplitude, and latency of the compound action potential (CAP). Midmodiolar sections of the implanted cochleas were analyzed in terms of trauma to hair cells, modiolar wall, osseous spiral lamina (OSL) and lateral wall. Results: Animals were assigned to cochlear trauma categories: minimal (n = 3), moderate (n = 5), or severe (n = 3). After cochleostomy and array insertion, CAP threshold shifts increased with trauma severity. At each stage a threshold shift at high frequencies (4-16 kHz) was accompanied with a threshold shift at low frequencies (0.25-2 kHz) that was 10-20 dB smaller. Withdrawal of the array led to a further worsening of responses, which probably indicates that insertion and removal trauma affected the responses rather than the mere presence of the array. In two instances, CAP threshold shifts were considerably larger than threshold shifts of cochlear microphonics, which could be explained by neural damage due to OSL fracture. A change in amplitudes at high sound levels was strongly correlated with threshold shifts, which is relevant for clinical ECochG performed at one sound level. Conclusion: Basal trauma caused by cochleostomy and/or array insertion should be minimized in order to preserve the low-frequency residual hearing of CI recipients.

4.
Hear Res ; 426: 108643, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36343534

RESUMO

Cochlear implants (CIs) provide acoustic information to implanted patients by electrically stimulating nearby auditory nerve fibers (ANFs) which then transmit the information to higher-level neural structures for further processing and interpretation. Computational models that simulate ANF responses to CI stimuli enable the exploration of the mechanisms underlying CI performance beyond the capacity of in vivo experimentation alone. However, all ANF models developed to date utilize to some extent anatomical/morphometric data, biophysical properties and/or physiological data measured in non-human animal models. This review compares response properties of the electrically stimulated auditory nerve (AN) in human listeners and different mammalian models. Properties of AN responses to single pulse stimulation, paired-pulse stimulation, and pulse-train stimulation are presented. While some AN response properties are similar between human listeners and animal models (e.g., increased AN sensitivity to single pulse stimuli with long interphase gaps), there are some significant differences. For example, the AN of most animal models is typically more sensitive to cathodic stimulation while the AN of human listeners is generally more sensitive to anodic stimulation. Additionally, there are substantial differences in the speed of recovery from neural adaptation between animal models and human listeners. Therefore, results from animal models cannot be simply translated to human listeners. Recognizing the differences in responses of the AN to electrical stimulation between humans and other mammals is an important step for creating ANF models that are more applicable to various human CI patient populations.


Assuntos
Implante Coclear , Implantes Cocleares , Animais , Humanos , Nervo Coclear/fisiologia , Estimulação Elétrica/métodos , Potenciais Evocados Auditivos , Mamíferos , Modelos Animais
5.
Biomedicines ; 10(11)2022 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-36428503

RESUMO

The mammalian target of rapamycin (mTOR) signaling plays a critical role in cell homeostasis, growth and survival. Here, we investigated the localization of the main mTOR signaling proteins in the organ of Corti of normal-hearing and deafened guinea pigs, as well as their possible modulation by exogenously administered brain-derived neurotrophic factor (BDNF) in deafened guinea pigs. Animals were ototoxically deafened by systemic administration of kanamycin and furosemide, and one week later, the right cochleas were treated with gelatin sponge soaked in rhBDNF, while the left cochleas were used as negative controls. Twenty-four hours after treatment, animals were euthanized, and the cochleas were processed for subsequent analysis. Through immunofluorescence, we demonstrated the localization of AKT, pAKT, mTOR, pmTOR and PTEN proteins throughout the cochlea of guinea pigs for the first time, with a higher expression in supporting cells. Moreover, an increase in mTOR immunostaining was observed in BDNF-treated cochleas by means of fluorescence intensity compared to the other groups. Conversely, Western blot analysis showed no significant differences in the protein levels between groups, probably due to dilution of proteins in the neighboring tissues of the organ of Corti. Altogether, our data indicate that mTOR signaling proteins are expressed by the organ of Corti (with a major role for supporting cells) and that the modulation of mTOR may be a protective mechanism triggered by BDNF in the degenerating organ of Corti.

6.
Front Mol Neurosci ; 15: 935111, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36226314

RESUMO

Severe hearing loss or deafness is often caused by cochlear hair cell loss and can be mitigated by a cochlear implant (CI). CIs target the auditory nerve, consisting of spiral ganglion cells (SGCs), which degenerate gradually, following hair cell loss. In animal models, it has been established that treatment with the neurotrophins brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3) reduce SGC degeneration. In this study, we aimed to investigate whether treatment with both BDNF and NT-3 (Cocktail) is superior to treatment with each neurotrophin separately regarding cell preservation and neural responsiveness to electrical stimulation. To this end, deafened guinea pigs received neurotrophic treatment in their right ear via a gelatin sponge on the perforated round window membrane, followed by cochlear implantation 4 weeks later in the same ear for electrophysiological recordings to various stimulation paradigms. Normal-hearing and deafened untreated guinea pigs were included as positive and negative controls, respectively. Substantial SGC loss occurred in all deafened animals. Each of the neurotrophic treatments led to enhanced SGC survival mainly in the basal turn of the cochlea, gradually decreasing toward the apex. The Cocktail treatment resulted in the highest SGC survival in the treated ear, followed by BDNF, with the least protection of SGCs following NT-3 treatment. Survival of the SGC's peripheral processes (PPs) followed the same trend in response to the treatment. However, survival of SGCs and PPs in the contralateral untreated ears was also highest in the Cocktail group. Consequently, analysis of the ratio between the treated and untreated ears showed that the BDNF group, which showed low SGC survival in the untreated ear, had the highest relative SGC survival of the three neurotrophin-treated groups. Neurotrophic treatment had positive effects in part of the electrically evoked compound action-potential recording paradigms. These effects were only observed for the BDNF or Cocktail treatment. We conclude that treatment with either BDNF or a cocktail of BDNF and NT-3 is preferred to NT-3 alone. Furthermore, since the Cocktail treatment resulted in better electrophysiological responsiveness and overall higher SGC survival than BDNF alone, we are inclined to recommend the Cocktail treatment rather than BDNF alone.

7.
J Assoc Res Otolaryngol ; 23(6): 721-738, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35948695

RESUMO

The electrically evoked compound action potential (eCAP) is a direct measure of the responsiveness of the auditory nerve to electrical stimulation from a cochlear implant (CI). CIs offer a unique opportunity to study the auditory nerve's electrophysiological behavior in individual human subjects over time. In order to understand exactly how the eCAP relates to the condition of the auditory nerve, it is crucial to compare changes in the eCAP over time in a controlled model of deafness-induced auditory nerve degeneration. In the present study, 10 normal-hearing young adult guinea pigs were implanted and deafened 4 weeks later, so that the effect of deafening could be monitored within-subject over time. Following implantation, but before deafening, most examined eCAP characteristics significantly changed, suggesting increasing excitation efficacy (e.g., higher maximum amplitude, lower threshold, shorter latency). Conversely, inter-phase gap (IPG) effects on these measures - within-subject difference measures that have been shown to correlate well with auditory nerve survival - did not vary for most eCAP characteristics. After deafening, we observed an initial increase in excitability (steeper slope of the eCAP amplitude growth function (AGF), lower threshold, shorter latency and peak width) which typically returned to normal-hearing levels within a week, after which a slower process, probably reflecting spiral ganglion cell loss, took place over the remaining 6 weeks (e.g., decrease in maximum amplitude, AGF slope, peak area, and IPG effect for AGF slope; increase in IPG effect for latency). Our results suggest that gradual changes in peak width and latency reflect the rate of neural degeneration, while peak area, maximum amplitude, and AGF slope reflect neural population size, which may be valuable for clinical diagnostics.


Assuntos
Implante Coclear , Implantes Cocleares , Adulto Jovem , Cobaias , Humanos , Animais , Potenciais de Ação/fisiologia , Potenciais Evocados , Nervo Coclear/fisiologia , Gânglio Espiral da Cóclea/fisiologia , Implante Coclear/métodos , Estimulação Elétrica , Potenciais Evocados Auditivos/fisiologia
8.
Ear Hear ; 43(1): 150-164, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34241983

RESUMO

OBJECTIVES: Amplitudes of electrically evoked compound action potentials (eCAPs) as a function of the stimulation level constitute the eCAP amplitude growth function (AGF). The slope of the eCAP AGF (i.e., rate of growth of eCAP amplitude as a function of stimulation level), recorded from subjects with cochlear implants (CIs), has been widely used as an indicator of survival of cochlear nerve fibers. However, substantial variation in the approach used to calculate the slope of the eCAP AGF makes it difficult to compare results across studies. In this study, we developed an improved slope-fitting method by addressing the limitations of previously used approaches and ensuring its application for the estimation of the maximum slopes of the eCAP AGFs recorded in both animal models and human listeners with various etiologies. DESIGN: The new eCAP AGF fitting method was designed based on sliding window linear regression. Slopes of the eCAP AGF estimated using this new fitting method were calculated and compared with those estimated using four other fitting methods reported in the literature. These four methods were nonlinear regression with a sigmoid function, linear regression, gradient calculation, and boxcar smoothing. The comparison was based on the fitting results of 72 eCAP AGFs recorded from 18 acutely implanted guinea pigs, 46 eCAP AGFs recorded from 23 chronically implanted guinea pigs, and 2094 eCAP AGFs recorded from 200 human CI users from 4 patient populations. The effect of the choice of input units of the eCAP AGF (linear versus logarithmic) on fitting results was also evaluated. RESULTS: The slope of the eCAP AGF was significantly influenced by the slope-fitting method and by the choice of input units. Overall, slopes estimated using all five fitting methods reflected known patterns of neural survival in human patient populations and were significantly correlated with speech perception scores. However, slopes estimated using the newly developed method showed the highest correlation with spiral ganglion neuron density among all five fitting methods for animal models. In addition, this new method could reliably and accurately estimate the slope for 4 human patient populations, while the performance of the other methods was highly influenced by the morphology of the eCAP AGF. CONCLUSIONS: The novel slope-fitting method presented in this study addressed the limitations of the other methods reported in the literature and successfully characterized the slope of the eCAP AGF for various animal models and CI patient populations. This method may be useful for researchers in conducting scientific studies and for clinicians in providing clinical care for CI users.


Assuntos
Implante Coclear , Implantes Cocleares , Potenciais de Ação/fisiologia , Animais , Nervo Coclear , Estimulação Elétrica , Potenciais Evocados Auditivos/fisiologia , Cobaias , Humanos
9.
Front Mol Neurosci ; 14: 729625, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34675775

RESUMO

Sensorineural hearing loss is mainly caused by irreversible damage to sensory hair cells (HCs). A subgroup of supporting cells (SCs) in the cochlea express leucine-rich repeat-containing G-protein coupled receptor 5 (LGR5), a marker for tissue-resident stem cells. LGR5+ SCs could be used as an endogenous source of stem cells for regeneration of HCs to treat hearing loss. Here, we report long-term presence of LGR5+ SCs in the mature adult cochlea and survival of LGR5+ SCs after severe ototoxic trauma characterized by partial loss of inner HCs and complete loss of outer HCs. Surviving LGR5+ SCs (confirmed by GFP expression) were located in the third row of Deiters' cells. We observed a change in the intracellular localization of GFP, from the nucleus in normal-hearing to cytoplasm and membrane in deafened mice. These data suggests that the adult mammalian cochlea possesses properties essential for regeneration even after severe ototoxic trauma.

10.
Brain Sci ; 12(1)2021 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-35053747

RESUMO

We investigated whether treatment with brain-derived neurotrophic factor (BDNF), which is known to protect spiral ganglion cells (SGCs), could also protect hair cells (HCs) and supporting cells (SCs) in the organ of Corti of a guinea pig model of sensorineural hearing loss. Hearing loss was induced by administration of kanamycin/furosemide and two BDNF treatments were performed: (1) by gelatin sponge (BDNF-GS) with acute cochlear implantation (CI), and (2) through a mini-osmotic pump (BDNF-OP) with chronic CI. Outer HCs (OHCs), inner HCs (IHCs), Border, Phalangeal, Pillar, Deiters', and Hensen's cells were counted. The BDNF-GS cochleas had significantly fewer OHCs compared to the untreated ones, while the IHC and SC numbers did not differ between treated and untreated cochleas. The BDNF-OP group showed similar cell numbers to the untreated group. SGC packing density was not correlated with the total number of SCs for either BDNF group. Our data suggest that: (1) BDNF does not prevent cell death in the organ of Corti, and that the protection of SGCs could result from a direct targeting by BDNF; (2) BDNF might induce a different function/activity of the remaining cells in the organ of Corti (independently from cell number).

11.
Hear Res ; 400: 108114, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33271438

RESUMO

Treatment with neurotrophins prevents degeneration of spiral ganglion cells (SGCs) after severe hair cell loss. In a previous study we demonstrated a long-lasting effect with brain-derived neurotrophic factor (BDNF) after cessation of treatment. In that study the survival of the SGC cell bodies was examined. Here we address the question whether their peripheral processes and central processes (axons) were protected by this treatment as well in the cochleas of the aforementioned study. Guinea pigs were deafened by co-administration of kanamycin and furosemide. Two weeks after deafening the right cochleas were implanted with an intracochlear electrode array combined with a cannula connected to an osmotic pump filled with BDNF solution. Four weeks later the treatment was stopped by surgically removing the osmotic pump. At that point, or another four or eight weeks later, the animals were sacrificed for histological analysis. Control groups consisted of normal-hearing animals, and three groups of deafened animals: two-weeks-deaf untreated animals, and six- and fourteen-weeks-deaf sham-treated animals. Cochleas were processed for analysis of: (1) the myelinated portion of peripheral processes in the osseous spiral lamina, (2) the cell bodies in Rosenthal's canal, and (3) axons in the internal acoustic meatus. Packing densities and cross-sectional areas were determined using light microscopy. Up to eight weeks after treatment cessation the numbers of peripheral processes and axons were significantly higher than in untreated cochleas of control animals. Whereas the numbers of cell bodies and axons were similar to those at the start of treatment, the peripheral processes were significantly less well preserved. This smaller protective effect was found mainly in the apical turns. Strategies to prevent SGC degeneration after hair cell loss should consider the differential effects on the various neural elements.


Assuntos
Surdez , Gânglio Espiral da Cóclea , Animais , Axônios , Fator Neurotrófico Derivado do Encéfalo , Corpo Celular , Sobrevivência Celular , Cóclea/patologia , Surdez/patologia , Cobaias , Degeneração Neural , Gânglio Espiral da Cóclea/patologia
12.
Brain Sci ; 10(11)2020 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-33126525

RESUMO

In deaf subjects using a cochlear implant (CI) for hearing restoration, the auditory nerve is subject to degeneration, which may negatively impact CI effectiveness. This nerve degeneration can be reduced by neurotrophic treatment. Here, we compare the preservative effects of the naturally occurring tyrosine receptor kinase B (TrkB) agonist brain-derived neurotrophic factor (BDNF) and the small-molecule TrkB agonist 7,8,3'-trihydroxyflavone (THF) on the auditory nerve in deafened guinea pigs. THF may be more effective than BDNF throughout the cochlea because of better pharmacokinetic properties. The neurotrophic compounds were delivered by placement of a gelatin sponge on the perforated round window membrane. To complement the histology of spiral ganglion cells (SGCs), electrically evoked compound action potential (eCAP) recordings were performed four weeks after treatment initiation. We analyzed the eCAP inter-phase gap (IPG) effect and measures derived from pulse-train evoked eCAPs, both indicative of SGC healthiness. BDNF but not THF yielded a significantly higher survival of SGCs in the basal cochlear turn than untreated controls. Regarding IPG effect and pulse-train responses, the BDNF-treated animals exhibited more normal responses than both untreated and THF-treated animals. We have thus confirmed the protective effect of BDNF, but we have not confirmed previously reported protective effects of THF with our clinically applicable delivery method.

13.
Hear Res ; 390: 107928, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32143110

RESUMO

Severe damage to the organ of Corti leads to degeneration of the spiral ganglion cells (SGCs) which form the auditory nerve. This degeneration starts at the level of synaptic connection of the peripheral processes (PPs) of SGCs with the cochlear hair cells. It is generally thought that from this point SGC degeneration progresses in a retrograde fashion: PPs degenerate first, followed by the SGC soma with a delay of several weeks to many months. Evidence for this course of events, both in animals and in humans, is not unambiguous, while this knowledge is important since the presence or absence of the different neural elements may greatly influence the response to electrical stimulation with a cochlear implant (CI). We therefore aimed to provide a comprehensive account of the course of SGC degeneration in the guinea pig cochlea after ototoxic treatment. Histological analysis of eighteen healthy and thirty-three deafened cochleas showed that the degeneration of SGCs and their peripheral processes was simultaneous rather than sequential. As the site of excitation for electrical stimulation with a CI may depend on the course of degeneration of the various neural elements, this finding is relevant both for understanding the electrophysiological mechanisms behind cochlear implantation and for recent efforts to induce PP resprouting for improved electrode-neural interface. Since excitation of the PPs is often thought to result in (secondary) longer-latency activity, we tested the hypothesis that having relatively many PPs produces a larger N2 peak in the electrically evoked compound action potential (eCAP); the present findings however do not support this theory. The course of the degeneration process may vary among species, and may depend on the cause of deafness, but the present findings at least indicate that gradual retrograde degeneration of the auditory nerve is not an elemental process following severe damage to the organ of Corti.


Assuntos
Surdez/patologia , Células Ciliadas Auditivas/ultraestrutura , Degeneração Neural , Gânglio Espiral da Cóclea/ultraestrutura , Animais , Limiar Auditivo , Surdez/induzido quimicamente , Surdez/fisiopatologia , Modelos Animais de Doenças , Potenciais Evocados Auditivos do Tronco Encefálico , Furosemida , Cobaias , Canamicina , Ototoxicidade , Gânglio Espiral da Cóclea/fisiopatologia
14.
Biomed Res Int ; 2018: 1568414, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30151372

RESUMO

Round window membrane (RWM) application of ouabain is known to selectively destroy type I spiral ganglion cells (SGCs) in cochleas of several rodent species, while leaving hair cells intact. This protocol has been used in rats and Mongolian gerbils, but observations in the guinea pig are conflicting. This is why we reinvestigated the effect of ouabain on the guinea pig cochlea. Ouabain solutions of different concentrations were placed, in a piece of gelfoam, upon the RWM of the right cochleas. Auditory function was assessed using acoustically evoked auditory brainstem responses (aABR). Finally, cochleas were fixed and processed for histological examination. Due to variability within treatment groups, histological data was pooled and three categories based upon general histological observations were defined: cochleas without outer hair cell (OHC) and SGC loss (Category 1), cochleas with OHC loss only (Category 2), and cochleas with OHC and SGC loss (Category 3). Animals treated with 1 mM or 10 mM ouabain showed shifts in hearing thresholds, corresponding with varying histological changes in their cochleas. Most cochleas exhibited complete outer hair cell loss in the basal and middle turns, while some had no changes, together with either moderate or near-complete loss of SGCs. Neither loss of inner hair cells nor histological changes of the stria vascularis were observed in any of the animals. Cochleas in Category 1 had normal aABRs and morphology. On average, in Category 2 OHC loss was 46.0±5.7%, SGC loss was below threshold, ABR threshold shift was 44.9±2.7 dB, and ABR wave II amplitude was decreased by 17.1±3.8 dB. In Category 3 OHC loss was 68.3±6.9%, SGC loss was 49.4±4.3%, ABR threshold shift was 39.0±2.4 dB, and ABR amplitude was decreased by 15.8±1.6 dB. Our results show that ouabain does not solely destroy type I SGCs in the guinea pig cochlea.


Assuntos
Inibidores Enzimáticos/toxicidade , Ouabaína/toxicidade , Gânglio Espiral da Cóclea/efeitos dos fármacos , Animais , Limiar Auditivo , Cóclea/efeitos dos fármacos , Potenciais Evocados Auditivos do Tronco Encefálico , Feminino , Cobaias , Células Ciliadas Auditivas Externas , Masculino
15.
Hear Res ; 345: 79-87, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28087418

RESUMO

Damage to and loss of the organ of Corti leads to secondary degeneration of the spiral ganglion cell (SGC) somata of the auditory nerve. Extensively examined in animal models, this degeneration process of SGC somata following deafening is well known. However, degeneration of auditory nerve axons, which conduct auditory information towards the brainstem, and its relation to SGC soma degeneration are largely unknown. The consequences of degeneration of the axons are relevant for cochlear implantation, which is applied to a deafened system but depends on the condition of the auditory nerve. We investigated the time sequence of degeneration of myelinated type I axons in deafened guinea pigs. Auditory nerves in six normal-hearing and twelve deafened animals, two, six and fourteen weeks (for each group four) after deafening were histologically analyzed. We developed a semi-automated method for axon counting, which allowed for a relatively large sample size (20% of the total cross-sectional area of the auditory nerve). We observed a substantial loss of auditory nerve area (29%), reduction in axon number (59%) and decrease in axoplasm area (41%) fourteen weeks after deafening compared to normal-hearing controls. The correlation between axonal degeneration and that of the SGC somata in the same cochleas was high, although axonal structures appeared to persist longer than the somata, suggesting a slower degeneration process. In the first two weeks after induction of deafness, the axonal cross-sectional area decreased but the axon number did not. In conclusion, the data strongly suggest that each surviving SGC possesses an axon.


Assuntos
Axônios/patologia , Nervo Coclear/patologia , Perda Auditiva Neurossensorial/patologia , Degeneração Neural , Gânglio Espiral da Cóclea/patologia , Animais , Nervo Coclear/fisiopatologia , Modelos Animais de Doenças , Feminino , Cobaias , Audição , Perda Auditiva Neurossensorial/fisiopatologia , Índice de Gravidade de Doença , Gânglio Espiral da Cóclea/fisiopatologia , Fatores de Tempo
16.
Int J Mol Sci ; 17(12)2016 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-27898033

RESUMO

Animal studies have evidenced protection of the auditory nerve by exogenous neurotrophic factors. In order to assess clinical applicability of neurotrophic treatment of the auditory nerve, the safety and efficacy of neurotrophic therapies in various human disorders were systematically reviewed. Outcomes of our literature search included disorder, neurotrophic factor, administration route, therapeutic outcome, and adverse event. From 2103 articles retrieved, 20 randomized controlled trials including 3974 patients were selected. Amyotrophic lateral sclerosis (53%) was the most frequently reported indication for neurotrophic therapy followed by diabetic polyneuropathy (28%). Ciliary neurotrophic factor (50%), nerve growth factor (24%) and insulin-like growth factor (21%) were most often used. Injection site reaction was a frequently occurring adverse event (61%) followed by asthenia (24%) and gastrointestinal disturbances (20%). Eighteen out of 20 trials deemed neurotrophic therapy to be safe, and six out of 17 studies concluded the neurotrophic therapy to be effective. Positive outcomes were generally small or contradicted by other studies. Most non-neurodegenerative diseases treated by targeted deliveries of neurotrophic factors were considered safe and effective. Hence, since local delivery to the cochlea is feasible, translation from animal studies to human trials in treating auditory nerve degeneration seems promising.


Assuntos
Nervo Coclear/efeitos dos fármacos , Fatores de Crescimento Neural/uso terapêutico , Animais , Humanos , Fatores de Crescimento Neural/administração & dosagem , Fatores de Crescimento Neural/efeitos adversos , Doenças Neurodegenerativas/tratamento farmacológico , Ensaios Clínicos Controlados Aleatórios como Assunto
17.
Adv Exp Med Biol ; 894: 143-153, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27080655

RESUMO

The electrically evoked compound action potential (eCAP) is a routinely performed measure of the auditory nerve in cochlear implant users. Using a convolution model of the eCAP, additional information about the neural firing properties can be obtained, which may provide relevant information about the health of the auditory nerve. In this study, guinea pigs with various degrees of nerve degeneration were used to directly relate firing properties to nerve histology. The same convolution model was applied on human eCAPs to examine similarities and ultimately to examine its clinical applicability. For most eCAPs, the estimated nerve firing probability was bimodal and could be parameterised by two Gaussian distributions with an average latency difference of 0.4 ms. The ratio of the scaling factors of the late and early component increased with neural degeneration in the guinea pig. This ratio decreased with stimulation intensity in humans. The latency of the early component decreased with neural degeneration in the guinea pig. Indirectly, this was observed in humans as well, assuming that the cochlear base exhibits more neural degeneration than the apex. Differences between guinea pigs and humans were observed, among other parameters, in the width of the early component: very robust in guinea pig, and dependent on stimulation intensity and cochlear region in humans. We conclude that the deconvolution of the eCAP is a valuable addition to existing analyses, in particular as it reveals two separate firing components in the auditory nerve.


Assuntos
Implantes Cocleares , Nervo Coclear/fisiologia , Potenciais de Ação/fisiologia , Animais , Estimulação Elétrica , Cobaias , Humanos
18.
J Neurosci ; 35(36): 12331-45, 2015 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-26354903

RESUMO

After substantial loss of cochlear hair cells, exogenous neurotrophins prevent degeneration of the auditory nerve. Because cochlear implantation, the current therapy for profound sensorineural hearing loss, depends on a functional nerve, application of neurotrophins is being investigated. We addressed two questions important for fundamental insight into the effects of exogenous neurotrophins on a degenerating neural system, and for translation to the clinic. First, does temporary treatment with brain-derived neurotrophic factor (BDNF) prevent nerve degeneration on the long term? Second, how does a BDNF-treated nerve respond to electrical stimulation? Deafened guinea pigs received a cochlear implant, and their cochleas were infused with BDNF for 4 weeks. Up to 8 weeks after treatment, their cochleas were analyzed histologically. Electrically evoked compound action potentials (eCAPs) were recorded using stimulation paradigms that are informative of neural survival. Spiral ganglion cell (SGC) degeneration was prevented during BDNF treatment, resulting in 1.9 times more SGCs than in deafened untreated cochleas. Importantly, SGC survival was almost complete 8 weeks after treatment cessation, when 2.6 times more SGCs were observed. In four eCAP characteristics (three involving alteration of the interphase gap of the biphasic current pulse and one involving pulse trains), we found large and statistically significant differences between normal-hearing and deaf controls. Importantly, for BDNF-treated animals, these eCAP characteristics were near normal, suggesting healthy responsiveness of BDNF-treated SGCs. In conclusion, clinically practicable short-term neurotrophin treatment is sufficient for long-term survival of SGCs, and it can restore or preserve SGC function well beyond the treatment period. Significance statement: Successful restoration of hearing in deaf subjects by means of a cochlear implant requires a healthy spiral ganglion cell population. Deafness-induced degeneration of these cells can be averted with neurotrophic factors. In the present study in deafened guinea pigs, we investigated the long-term effects of temporary (i.e., clinically practicable) treatment with brain-derived neurotrophic factor (BDNF). We show that, after treatment cessation, the neuroprotective effect remains for at least 8 weeks. Moreover, for the first time, it is shown that the electrical responsiveness of BDNF-treated spiral ganglion cells is preserved during this period as well. These findings demonstrate that treatment of the auditory nerve with neurotrophic factors may be relevant for cochlear implant users.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/farmacologia , Nervo Coclear/efeitos dos fármacos , Surdez/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Potenciais de Ação , Animais , Fator Neurotrófico Derivado do Encéfalo/uso terapêutico , Nervo Coclear/patologia , Nervo Coclear/fisiologia , Surdez/patologia , Feminino , Cobaias , Fármacos Neuroprotetores/uso terapêutico , Gânglio Espiral da Cóclea/citologia , Gânglio Espiral da Cóclea/efeitos dos fármacos , Gânglio Espiral da Cóclea/fisiologia
19.
Hear Res ; 321: 12-24, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25582354

RESUMO

Successful cochlear implant performance requires adequate responsiveness of the auditory nerve to prolonged pulsatile electrical stimulation. Degeneration of the auditory nerve as a result of severe hair cell loss could considerably compromise this ability. The main objective of this study was to characterize the recovery of the electrically stimulated auditory nerve, as well as to evaluate possible changes caused by deafness-induced degeneration. To this end we studied temporal responsiveness of the auditory nerve in a guinea pig model of sensorineural hearing loss. Using masker-probe and pulse train paradigms we compared electrically evoked compound action potentials (eCAPs) in normal-hearing animals with those in animals with moderate (two weeks after ototoxic treatment) and severe (six weeks after ototoxic treatment) loss of spiral ganglion cells (SGCs). Masker-probe interval and pulse train inter-pulse interval was varied from 0.3 to 16 ms. Whereas recovery assessed with masker-probe was roughly similar for normal-hearing and both groups of deafened animals, it was considerably faster for six weeks deaf animals (τ ≈ 1.2 ms) than for two weeks deaf or normal-hearing animals (τ ≈ 3-4 ms) when 100-ms pulse trains were applied. Latency increased with decreasing inter-pulse intervals, and this was more pronounced with pulse trains than with masker-probe stimulation. With high frequency pulse train stimulation eCAP amplitudes were modulated for deafened animals, meaning that amplitudes for odd pulse numbers were larger than for even pulses. The relative refractory period (τ) and the modulation depth of the eCAP amplitude for pulse trains, as well as the latency increase for both paradigms significantly correlated with quantified measures of auditory nerve degeneration (size and packing density of SGCs). In addition to these findings, separate masker-probe recovery functions for the eCAP N1 and N2 peaks displayed a robust non-monotonic or shoulder-shaped course in all animals. The time interval between the N1 and N2 correlated with neuronal refractoriness, suggesting that the N2 peak reflects a second firing of part of the SGC population. We conclude that - compared to the commonly used masker-probe recovery functions - recovery functions obtained with pulse train stimulation may provide a means to augment differences and, by doing so, to more potently discriminate between auditory nerve conditions.


Assuntos
Implante Coclear , Nervo Coclear/fisiopatologia , Perda Auditiva Neurossensorial/fisiopatologia , Animais , Percepção Auditiva , Nervo Coclear/patologia , Modelos Animais de Doenças , Estimulação Elétrica , Potenciais Evocados Auditivos , Feminino , Furosemida , Cobaias , Audição , Perda Auditiva Neurossensorial/induzido quimicamente , Perda Auditiva Neurossensorial/patologia , Canamicina , Degeneração Neural , Mascaramento Perceptivo , Tempo de Reação , Recuperação de Função Fisiológica , Período Refratário Eletrofisiológico , Gânglio Espiral da Cóclea/fisiopatologia , Fatores de Tempo
20.
J Assoc Res Otolaryngol ; 15(2): 187-202, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24469861

RESUMO

After severe hair cell loss, secondary degeneration of spiral ganglion cells (SGCs) is observed-a gradual process that spans years in humans but only takes weeks in guinea pigs. Being the target for cochlear implants (CIs), the physiological state of the SGCs is important for the effectiveness of a CI. For assessment of the nerve's state, focus has generally been on its response threshold. Our goal was to add a more detailed characterization of SGC functionality. To this end, the electrically evoked compound action potential (eCAP) was recorded in normal-hearing guinea pigs and guinea pigs that were deafened 2 or 6 weeks prior to the experiments. We evaluated changes in eCAP characteristics when the phase duration (PD) and inter-phase gap (IPG) of a biphasic current pulse were varied. We correlated the magnitude of these changes to quantified histological measures of neurodegeneration (SGC packing density and SGC size). The maximum eCAP amplitude, derived from the input-output function, decreased after deafening, and increased with both PD and IPG. The eCAP threshold did not change after deafening, and decreased with increasing PD and IPG. The dynamic range was wider for the 6-weeks-deaf animals than for the other two groups. Excitability increased with IPG (steeper slope of the input-output function and lower stimulation level at the half-maximum eCAP amplitude), but to a lesser extent for the deafened animals than for normal-hearing controls. The latency was shorter for the 6-weeks-deaf animals than for the other two groups. For several of these eCAP characteristics, the effect size of IPG correlated well with histological measures of degeneration, whereas effect size of PD did not. These correlations depend on the use of high current levels, which could limit clinical application. Nevertheless, their potential of these correlations towards assessment of the condition of the auditory nerve may be of great benefit to clinical diagnostics and prognosis in cochlear implant recipients.


Assuntos
Nervo Coclear/fisiologia , Degeneração Neural/fisiopatologia , Potenciais de Ação , Animais , Surdez/fisiopatologia , Estimulação Elétrica , Feminino , Cobaias , Gânglio Espiral da Cóclea/citologia , Gânglio Espiral da Cóclea/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...