Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Front Mol Neurosci ; 11: 325, 2018.
Article in English | MEDLINE | ID: mdl-30319348

ABSTRACT

Bdnf exon-IV and exon-VI transcripts are driven by neuronal activity and are involved in pathologies related to sleep, fear or memory disorders. However, how their differential transcription translates activity changes into long-lasting network changes is elusive. Aiming to trace specifically the network controlled by exon-IV and -VI derived BDNF during activity-dependent plasticity changes, we generated a transgenic reporter mouse for B DNF- l ive- e xon- v isualization (BLEV), in which expression of Bdnf exon-IV and -VI can be visualized by co-expression of CFP and YFP. CFP and YFP expression was differentially activated and targeted in cell lines, primary cultures and BLEV reporter mice without interfering with BDNF protein synthesis. CFP and YFP expression, moreover, overlapped with BDNF protein expression in defined hippocampal neuronal, glial and vascular locations in vivo. So far, activity-dependent BDNF cannot be explicitly monitored independent of basal BDNF levels. The BLEV reporter mouse therefore provides a new model, which can be used to test whether stimulus-induced activity-dependent changes in BDNF expression are instrumental for long-lasting plasticity modifications.

2.
Front Mol Neurosci ; 11: 260, 2018.
Article in English | MEDLINE | ID: mdl-30127717

ABSTRACT

Activity-dependent BDNF (brain-derived neurotrophic factor) expression is hypothesized to be a cue for the context-specificity of memory formation. So far, activity-dependent BDNF cannot be explicitly monitored independently of basal BDNF levels. We used the BLEV ( B DNF- live-exon- visualization) reporter mouse to specifically detect activity-dependent usage of Bdnf exon-IV and -VI promoters through bi-cistronic co-expression of CFP and YFP, respectively. Enriching acoustic stimuli led to improved peripheral and central auditory brainstem responses, increased Schaffer collateral LTP, and enhanced performance in the Morris water maze. Within the brainstem, neuronal activity was increased and accompanied by a trend for higher expression levels of Bdnf exon-IV-CFP and exon-VI-YFP transcripts. In the hippocampus BDNF transcripts were clearly increased parallel to changes in parvalbumin expression and were localized to specific neurons and capillaries. Severe acoustic trauma, in contrast, elevated neither Bdnf transcript levels, nor auditory responses, parvalbumin or LTP. Together, this suggests that critical sensory input is essential for recruitment of activity-dependent auditory-specific BDNF expression that may shape network adaptation.

3.
Cell Physiol Biochem ; 47(4): 1509-1532, 2018.
Article in English | MEDLINE | ID: mdl-29940568

ABSTRACT

BACKGROUND/AIMS: From invertebrates to mammals, Gαi proteins act together with their common binding partner Gpsm2 to govern cell polarization and planar organization in virtually any polarized cell. Recently, we demonstrated that Gαi3-deficiency in pre-hearing murine cochleae pointed to a role of Gαi3 for asymmetric migration of the kinocilium as well as the orientation and shape of the stereociliary ("hair") bundle, a requirement for the progression of mature hearing. We found that the lack of Gαi3 impairs stereociliary elongation and hair bundle shape in high-frequency cochlear regions, linked to elevated hearing thresholds for high-frequency sound. How these morphological defects translate into hearing phenotypes is not clear. METHODS: Here, we studied global and conditional Gnai3 and Gnai2 mouse mutants deficient for either one or both Gαi proteins. Comparative analyses of global versus Foxg1-driven conditional mutants that mainly delete in the inner ear and telencephalon in combination with functional tests were applied to dissect essential and redundant functions of different Gαi isoforms and to assign specific defects to outer or inner hair cells, the auditory nerve, satellite cells or central auditory neurons. RESULTS: Here we report that lack of Gαi3 but not of the ubiquitously expressed Gαi2 elevates hearing threshold, accompanied by impaired hair bundle elongation and shape in high-frequency cochlear regions. During the crucial reprogramming of the immature inner hair cell (IHC) synapse into a functional sensory synapse of the mature IHC deficiency for Gαi2 or Gαi3 had no impact. In contrast, double-deficiency for Gαi2 and Gαi3 isoforms results in abnormalities along the entire tonotopic axis including profound deafness associated with stereocilia defects. In these mice, postnatal IHC synapse maturation is also impaired. In addition, the analysis of conditional versus global Gαi3-deficient mice revealed that the amplitude of ABR wave IV was disproportionally elevated in comparison to ABR wave I indicating that Gαi3 is selectively involved in generation of neural gain during auditory processing. CONCLUSION: We propose a so far unrecognized complexity of isoform-specific and overlapping Gαi protein functions particular during final differentiation processes.


Subject(s)
Carrier Proteins/metabolism , Forkhead Transcription Factors/metabolism , GTP-Binding Protein alpha Subunit, Gi2/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Hair Cells, Auditory, Inner/metabolism , Hearing/physiology , Nerve Tissue Proteins/metabolism , Animals , Carrier Proteins/genetics , Cell Cycle Proteins , Forkhead Transcription Factors/genetics , GTP-Binding Protein alpha Subunit, Gi2/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Hair Cells, Auditory, Inner/cytology , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics
4.
FASEB J ; 32(6): 3005-3019, 2018 06.
Article in English | MEDLINE | ID: mdl-29401591

ABSTRACT

Systemic corticosteroids have been the mainstay of treatment for various hearing disorders for more than 30 yr. Accordingly, numerous studies have described glucocorticoids (GCs) and stressors to be protective in the auditory organ against damage associated with a variety of health conditions, including noise exposure. Conversely, stressors are also predictive risk factors for hearing disorders. How both of these contrasting stress actions are linked has remained elusive. Here, we demonstrate that higher corticosterone levels during acoustic trauma in female rats is highly correlated with a decline of auditory fiber responses in high-frequency cochlear regions, and that hearing thresholds and the outer hair cell functions (distortion products of otoacoustic emissions) are left unaffected. Moreover, when GC receptor (GR) or mineralocorticoid receptor (MR) activation was antagonized by mifepristone or spironolactone, respectively, GR, but not MR, inhibition significantly and permanently attenuated trauma-induced effects on auditory fiber responses, including inner hair cell ribbon loss and related reductions of early and late auditory brainstem responses. These findings strongly imply that higher corticosterone stress levels profoundly impair auditory nerve processing, which may influence central auditory acuity. These changes are likely GR mediated as they are prevented by mifepristone.-Singer, W., Kasini, K., Manthey, M., Eckert, P., Armbruster, P., Vogt, M. A., Jaumann, M., Dotta, M., Yamahara, K., Harasztosi, C., Zimmermann, U., Knipper, M., Rüttiger, L. The glucocorticoid antagonist mifepristone attenuates sound-induced long-term deficits in auditory nerve response and central auditory processing in female rats.


Subject(s)
Cochlear Nerve/physiopathology , Evoked Potentials, Auditory, Brain Stem/drug effects , Glucocorticoids/antagonists & inhibitors , Hearing Disorders/physiopathology , Hearing Loss, Noise-Induced/physiopathology , Mifepristone/pharmacology , Animals , Cochlea/metabolism , Cochlea/pathology , Cochlea/physiopathology , Cochlear Nerve/metabolism , Cochlear Nerve/pathology , Female , Glucocorticoids/adverse effects , Glucocorticoids/pharmacology , Hearing Disorders/chemically induced , Hearing Disorders/drug therapy , Hearing Disorders/metabolism , Hearing Loss, Noise-Induced/chemically induced , Hearing Loss, Noise-Induced/drug therapy , Hearing Loss, Noise-Induced/metabolism , Rats , Rats, Wistar , Receptors, Glucocorticoid/metabolism , Receptors, Mineralocorticoid/metabolism
5.
Mol Pharmacol ; 92(4): 375-388, 2017 10.
Article in English | MEDLINE | ID: mdl-28874607

ABSTRACT

Nitric oxide (NO) activates the NO-sensitive soluble guanylate cyclase (NO-GC, sGC) and triggers intracellular signaling pathways involving cGMP. For survival of cochlear hair cells and preservation of hearing, NO-mediated cascades have both protective and detrimental potential. Here we examine the cochlear function of mice lacking one of the two NO-sensitive guanylate cyclase isoforms [NO-GC1 knockout (KO) or NO-GC2 KO]. The deletion of NO-GC1 or NO-GC2 did not influence electromechanical outer hair cell (OHC) properties, as measured by distortion product otoacoustic emissions, neither before nor after noise exposure, nor were click- or noise-burst-evoked auditory brainstem response thresholds different from controls. Yet inner hair cell (IHC) ribbons and auditory nerve responses showed significantly less deterioration in NO-GC1 KO and NO-GC2 KO mice after noise exposure. Consistent with a selective role of NO-GC in IHCs, NO-GC ß1 mRNA was found in isolated IHCs but not in OHCs. Using transgenic mice expressing the fluorescence resonance energy transfer-based cGMP biosensor cGi500, NO-induced elevation of cGMP was detected in real-time in IHCs but not in OHCs. Pharmacologic long-term treatment with a NO-GC stimulator altered auditory nerve responses but did not affect OHC function and hearing thresholds. Interestingly, NO-GC stimulation exacerbated the loss of auditory nerve response in aged animals but attenuated the loss in younger animals. We propose NO-GC2 and, to some degree, NO-GC1 as targets for early pharmacologic prevention of auditory fiber loss (synaptopathy). Both isoforms provide selective benefits for hearing function by maintaining the functional integrity of auditory nerve fibers in early life rather than at old age.


Subject(s)
Guanylate Cyclase/metabolism , Hair Cells, Auditory, Inner/metabolism , Hair Cells, Auditory, Inner/pathology , Nitric Oxide/metabolism , Noise/adverse effects , Receptors, Cell Surface/metabolism , Animals , Female , Hair Cells, Auditory, Inner/drug effects , Isoenzymes/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , Morpholines/pharmacology , Pyrimidines/pharmacology , Rats , Rats, Wistar , Receptors, Cell Surface/agonists , Synapses/drug effects , Synapses/metabolism , Synapses/pathology
6.
ORL J Otorhinolaryngol Relat Spec ; 79(1-2): 93-111, 2017.
Article in English | MEDLINE | ID: mdl-28231578

ABSTRACT

In medicine, biomarkers are a metric for disease state. More generally, a biomarker is anything that can be used as an indicator for a particular disease state or any physiological state of an organism. Here, we introduce functional and molecular biomarkers that are useful for categorizing defined subtypes of hearing disorder, which can help to selectively trace a particular dysfunction of the inner ear and the auditory pathway to disease.


Subject(s)
Biomarkers/blood , Hearing Disorders/blood , Hearing Disorders/diagnosis , Animals , Audiometry , Evoked Potentials, Auditory, Brain Stem , Female , Humans , Magnetic Resonance Imaging/methods , Male , Otoacoustic Emissions, Spontaneous , Predictive Value of Tests , Sensitivity and Specificity , Severity of Illness Index
7.
Neurobiol Aging ; 44: 173-184, 2016 08.
Article in English | MEDLINE | ID: mdl-27318145

ABSTRACT

A dramatic shift in societal demographics will lead to rapid growth in the number of older people with hearing deficits. Poorer performance in suprathreshold speech understanding and temporal processing with age has been previously linked with progressing inner hair cell (IHC) synaptopathy that precedes age-dependent elevation of auditory thresholds. We compared central sound responsiveness after acoustic trauma in young, middle-aged, and older rats. We demonstrate that IHC synaptopathy progresses from middle age onward and hearing threshold becomes elevated from old age onward. Interestingly, middle-aged animals could centrally compensate for the loss of auditory fiber activity through an increase in late auditory brainstem responses (late auditory brainstem response wave) linked to shortening of central response latencies. In contrast, old animals failed to restore central responsiveness, which correlated with reduced temporal resolution in responding to amplitude changes. These findings may suggest that cochlear IHC synaptopathy with age does not necessarily induce temporal auditory coding deficits, as long as the capacity to generate neuronal gain maintains normal sound-induced central amplitudes.


Subject(s)
Aging/physiology , Hair Cells, Auditory, Inner/physiology , Hearing Loss, Noise-Induced/physiopathology , Hearing/physiology , Animals , Auditory Perception/physiology , Auditory Threshold , Evoked Potentials, Auditory, Brain Stem/physiology , Female , Humans , Rats, Wistar , Reaction Time/physiology
8.
Mol Neurobiol ; 53(8): 5607-27, 2016 10.
Article in English | MEDLINE | ID: mdl-26476841

ABSTRACT

For all sensory organs, the establishment of spatial and temporal cortical resolution is assumed to be initiated by the first sensory experience and a BDNF-dependent increase in intracortical inhibition. To address the potential of cortical BDNF for sound processing, we used mice with a conditional deletion of BDNF in which Cre expression was under the control of the Pax2 or TrkC promoter. BDNF deletion profiles between these mice differ in the organ of Corti (BDNF (Pax2) -KO) versus the auditory cortex and hippocampus (BDNF (TrkC) -KO). We demonstrate that BDNF (Pax2) -KO but not BDNF (TrkC) -KO mice exhibit reduced sound-evoked suprathreshold ABR waves at the level of the auditory nerve (wave I) and inferior colliculus (IC) (wave IV), indicating that BDNF in lower brain regions but not in the auditory cortex improves sound sensitivity during hearing onset. Extracellular recording of IC neurons of BDNF (Pax2) mutant mice revealed that the reduced sensitivity of auditory fibers in these mice went hand in hand with elevated thresholds, reduced dynamic range, prolonged latency, and increased inhibitory strength in IC neurons. Reduced parvalbumin-positive contacts were found in the ascending auditory circuit, including the auditory cortex and hippocampus of BDNF (Pax2) -KO, but not of BDNF (TrkC) -KO mice. Also, BDNF (Pax2) -WT but not BDNF (Pax2) -KO mice did lose basal inhibitory strength in IC neurons after acoustic trauma. These findings suggest that BDNF in the lower parts of the auditory system drives auditory fidelity along the entire ascending pathway up to the cortex by increasing inhibitory strength in behaviorally relevant frequency regions. Fidelity and inhibitory strength can be lost following auditory nerve injury leading to diminished sensory outcome and increased central noise.


Subject(s)
Auditory Cortex/pathology , Auditory Cortex/physiopathology , Brain-Derived Neurotrophic Factor/metabolism , Noise , Animals , Auditory Cortex/metabolism , Auditory Threshold , Cochlea/metabolism , Evoked Potentials, Auditory, Brain Stem , Gene Deletion , Hearing , Inferior Colliculi/pathology , Inferior Colliculi/physiopathology , Integrases/metabolism , Mice, Knockout , Promoter Regions, Genetic/genetics , Receptor, trkC/metabolism , Risk Factors
9.
Cell Mol Life Sci ; 72(20): 3953-69, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25939269

ABSTRACT

The unconventional myosin VI, a member of the actin-based motor protein family of myosins, is expressed in the retina. Its deletion was previously shown to reduce amplitudes of the a- and b-waves of the electroretinogram. Analyzing wild-type and myosin VI-deficient Snell's Waltzer mice in more detail, the expression pattern of myosin VI in retinal pigment epithelium, outer limiting membrane, and outer plexiform layer could be linked with differential progressing ocular deficits. These encompassed reduced a-waves and b-waves and disturbed oscillatory potentials in the electroretinogram, photoreceptor cell death, retinal microglia infiltration, and formation of basal laminar deposits. A phenotype comprising features of glaucoma (neurodegeneration) and age-related macular degeneration could thus be uncovered that suggests dysfunction of myosin VI and its variable cargo adaptor proteins for membrane sorting and autophagy, as possible candidate mediators for both disease forms.


Subject(s)
Gene Deletion , Macular Degeneration/genetics , Myosin Heavy Chains/physiology , Optic Nerve Diseases/genetics , Animals , Genotype , Macular Degeneration/pathology , Mice , Mice, Inbred C57BL , Microglia/pathology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Optic Nerve Diseases/pathology , Photoreceptor Cells, Vertebrate/pathology , Retina/metabolism , Retina/physiology
10.
Cell Tissue Res ; 361(1): 77-93, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25843689

ABSTRACT

Before hearing onset, inner hair cell (IHC) maturation proceeds under the influence of spontaneous Ca(2+) action potentials (APs). The temporal signature of the IHC Ca(2+) AP is modified through an efferent cholinergic feedback from the medial olivocochlear bundle (MOC) and drives the IHC pre- and post-synapse phenotype towards low spontaneous (spike) rate (SR), high-threshold characteristics. With sensory experience, the IHC pre- and post-synapse phenotype matures towards the instruction of low-SR, high-threshold and of high-SR, low-threshold auditory fiber characteristics. Corticosteroid feedback together with local brain-derived nerve growth factor (BDNF) and catecholaminergic neurotransmitters (dopamine) might be essential for this developmental step. In this review, we address the question of whether the control of low-SR and high-SR fiber characteristics is linked to various degrees of vulnerability of auditory fibers in the mature system. In particular, we examine several IHC synaptopathies in the context of various hearing disorders and exemplified shortfalls before and after hearing onset.


Subject(s)
Cochlea/growth & development , Hair Cells, Auditory, Inner/metabolism , Hearing Disorders/genetics , Hearing Loss, Central/genetics , Hair Cells, Auditory, Inner/cytology , Humans
11.
Cell Physiol Biochem ; 35(5): 1905-23, 2015.
Article in English | MEDLINE | ID: mdl-25871611

ABSTRACT

BACKGROUND: Accumulating evidence suggests that tinnitus may occur despite normal auditory sensitivity, probably linked to partial degeneration of the cochlear nerve and damage of the inner hair cell (IHC) synapse. Damage to the IHC synapses and deafferentation may occur even after moderate noise exposure. For both salicylate- and noise-induced tinnitus, aberrant N-methyl-d-aspartate (NMDA) receptor activation and related auditory nerve excitation have been suggested as origin of cochlear tinnitus. Accordingly, NMDA receptor inhibition has been proposed as a pharmacologic approach for treatment of synaptopathic tinnitus. METHODS: Round-window application of the NMDA receptor antagonist AM-101 (Esketamine hydrochloride gel; Auris Medical AG, Basel, Switzerland) was tested in an animal model of tinnitus induced by acute traumatic noise. The study included the quantification of IHC ribbon synapses as a correlate for deafferentation as well as the measurement of the auditory brainstem response (ABR) to close-threshold sensation level stimuli as an indication of sound-induced auditory nerve activity. RESULTS: We have shown that AM-101 reduced the trauma-induced loss of IHC ribbons and counteracted the decline of ABR wave I amplitude generated in the cochlea/auditory nerve. CONCLUSION: Local round-window application of AM-101 may be a promising therapeutic intervention for the treatment of synaptopathic tinnitus.


Subject(s)
Cochlea/metabolism , Noise , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Anesthesia , Animals , Apoptosis Regulatory Proteins/therapeutic use , Apoptosis Regulatory Proteins/toxicity , Auditory Threshold/drug effects , Behavior, Animal/drug effects , Cochlea/drug effects , Disease Models, Animal , Female , Rats , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/drug effects , Synapses/metabolism , Tinnitus/drug therapy , Tinnitus/etiology
12.
Orphanet J Rare Dis ; 10: 15, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25759012

ABSTRACT

BACKGROUND: Early-onset hearing loss is mostly of genetic origin. The complexity of the hearing process is reflected by its extensive genetic heterogeneity, with probably many causative genes remaining to be identified. Here, we aimed at identifying the genetic basis for autosomal dominant non-syndromic hearing loss (ADNSHL) in a large German family. METHODS: A panel of 66 known deafness genes was analyzed for mutations by next-generation sequencing (NGS) in the index patient. We then conducted genome-wide linkage analysis, and whole-exome sequencing was carried out with samples of two patients. Expression of Osbpl2 in the mouse cochlea was determined by immunohistochemistry. Because Osbpl2 has been proposed as a target of miR-96, we investigated homozygous Mir96 mutant mice for its upregulation. RESULTS: Onset of hearing loss in the investigated ADNSHL family is in childhood, initially affecting the high frequencies and progressing to profound deafness in adulthood. However, there is considerable intrafamilial variability. We mapped a novel ADNSHL locus, DFNA67, to chromosome 20q13.2-q13.33, and subsequently identified a co-segregating heterozygous frameshift mutation, c.141_142delTG (p.Arg50Alafs*103), in OSBPL2, encoding a protein known to interact with the DFNA1 protein, DIAPH1. In mice, Osbpl2 was prominently expressed in stereocilia of cochlear outer and inner hair cells. We found no significant Osbpl2 upregulation at the mRNA level in homozygous Mir96 mutant mice. CONCLUSION: The function of OSBPL2 in the hearing process remains to be determined. Our study and the recent description of another frameshift mutation in a Chinese ADNSHL family identify OSBPL2 as a novel gene for progressive deafness.


Subject(s)
Deafness/genetics , Hair Cells, Auditory/metabolism , Receptors, Steroid/metabolism , Stereocilia/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Child, Preschool , Female , Gene Expression Regulation , Genetic Linkage , Humans , Infant , Male , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Pedigree , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Steroid/genetics , Young Adult
13.
Mol Cell Endocrinol ; 382(1): 26-37, 2014 Jan 25.
Article in English | MEDLINE | ID: mdl-24012852

ABSTRACT

Thyroid hormone acts on gene transcription by binding to its nuclear receptors TRα1 and TRß. Whereas global deletion of TRß causes deafness, global TRα-deficient mice have normal hearing thresholds. Since the individual roles of the two receptors in cochlear hair cells are still unclear, we generated mice with a hair cell-specific mutation of TRα1 or deletion of TRß using the Cre-loxP system. Hair cell-specific TRß mutant mice showed normal hearing thresholds but delayed BK channel expression in inner hair cells, slightly stronger outer hair cell function, and slightly reduced amplitudes of auditory brainstem responses. In contrast, hair cell-specific TRα mutant mice showed normal timing of BK channel expression, slightly reduced outer hair cell function, and slightly enhanced amplitudes of auditory brainstem responses. Our data demonstrate that TRß-related deafness originates outside of hair cells and that TRα and TRß play opposing, non-redundant roles in hair cells. A role for thyroid hormone receptors in controlling key regulators that shape signal transduction during development is discussed. Thyroid hormone may act through different thyroid hormone receptor activities to permanently alter the sensitivity of auditory neurotransmission.


Subject(s)
Hair Cells, Auditory/metabolism , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormone Receptors beta/metabolism , Afferent Pathways/metabolism , Animals , Auditory Perception , Brain Stem/metabolism , Gene Expression Regulation , Hair Cells, Auditory/physiology , KCNQ Potassium Channels/metabolism , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Mice , Mice, Knockout , Molecular Motor Proteins/metabolism , Organ Specificity , Otoacoustic Emissions, Spontaneous , Phenotype , Recombination, Genetic/genetics , Signal Transduction , Tectorial Membrane/growth & development , Tectorial Membrane/metabolism , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors beta/genetics
14.
Int J Infect Dis ; 18: 87-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24161208

ABSTRACT

We report a case of cryptococcal immune reconstitution inflammatory syndrome affecting the lungs, and 10 months later the cervical lymph nodes, in the absence of cryptococcal meningitis, in advanced HIV infection. Our report demonstrates the organ-specificity of the timing of the inflammatory response and illustrates the organ-specific interplay of immunity and infection in cryptococcal disease.


Subject(s)
HIV Infections/microbiology , Immune Reconstitution Inflammatory Syndrome/drug therapy , Meningitis, Cryptococcal/drug therapy , Adult , Antifungal Agents/therapeutic use , Cryptococcus/drug effects , DNA, Fungal/isolation & purification , Follow-Up Studies , HIV Infections/drug therapy , Humans , Lung/microbiology , Lymph Nodes/microbiology , Male , Viral Load
15.
Prog Neurobiol ; 111: 17-33, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24012803

ABSTRACT

The prevalence of hearing problems in the Western world has, due to aging of the population, doubled over the past 30 years. Thereby, noise-induced hearing loss is an important factor that worsens over time in addition to age-related hearing loss. Hearing loss is usually measured as an elevation of a person's hearing thresholds, expressed in decibel (dB). However, recent animal studies have unraveled a type of permanent cochlear damage, without an elevation of hearing thresholds. This subtle damage is linked to a permanent and progressive degeneration of auditory fibers that occurs in association with damage of the inner hair cell synapse. Afferent neuronal degeneration has been suggested to be involved in hyperacusis (over sensitivity to sound) and tinnitus (a phantom sound percept). Hyperacusis and tinnitus are potentially devastating conditions that are still incurable. The main risk factors to develop tinnitus or hyperacusis are hearing loss, social stress and age. Both tinnitus and hyperacusis have been discussed in the context of a pathological increased response gain in subcortical brain regions as a reaction to deprivation of sensory input. Novel studies confirm the involvement of peripheral deafferentation for tinnitus and hyperacusis, but suggest that the disorder results from different brain responses to different degrees of deafferentation: while tinnitus may arise as a failure of the brain to adapt to deprived peripheral input, hyperacusis may result from an 'over-adaptive' increase in response gain. Moreover, moderate and high stress levels at the time of acoustic trauma have been suggested to play a pivotal role in the vulnerability of the cochlea to acoustic damage and therefore for the development of tinnitus and hyperacusis.


Subject(s)
Cochlea/physiopathology , Hyperacusis/physiopathology , Neurobiology , Tinnitus/physiopathology , Animals , Auditory Threshold/physiology , Cochlea/pathology , Humans
16.
J Neurosci ; 33(22): 9508-19, 2013 May 29.
Article in English | MEDLINE | ID: mdl-23719817

ABSTRACT

The encoding of auditory information with indefatigable precision requires efficient resupply of vesicles at inner hair cell (IHC) ribbon synapses. Otoferlin, a transmembrane protein responsible for deafness in DFNB9 families, has been postulated to act as a calcium sensor for exocytosis as well as to be involved in rapid vesicle replenishment of IHCs. However, the molecular basis of vesicle recycling in IHCs is largely unknown. In the present study, we used high-resolution liquid chromatography coupled with mass spectrometry to copurify otoferlin interaction partners in the mammalian cochlea. We identified multiple subunits of the adaptor protein complex AP-2 (CLAP), an essential component of clathrin-mediated endocytosis, as binding partners of otoferlin in rats and mice. The interaction between otoferlin and AP-2 was confirmed by coimmunoprecipitation. We also found that AP-2 interacts with myosin VI, another otoferlin binding partner important for clathrin-mediated endocytosis (CME). The expression of AP-2 in IHCs was verified by reverse transcription PCR. Confocal microscopy experiments revealed that the expression of AP-2 and its colocalization with otoferlin is confined to mature IHCs. When CME was inhibited by blocking dynamin action, real-time changes in membrane capacitance showed impaired synaptic vesicle replenishment in mature but not immature IHCs. We suggest that an otoferlin-AP-2 interaction drives Ca(2+)- and stimulus-dependent compensating CME in mature IHCs.


Subject(s)
Clathrin/physiology , Cochlea/physiology , Endocytosis/physiology , Hair Cells, Auditory, Inner/physiology , Membrane Proteins/physiology , Adaptor Protein Complex 2/physiology , Animals , Cell Membrane/physiology , Cochlea/cytology , Electrophysiological Phenomena , Immunohistochemistry , Immunoprecipitation , Mass Spectrometry , Mice , Microscopy, Confocal , Myosin Heavy Chains/physiology , Protein Binding , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Synapses/physiology
17.
Histochem Cell Biol ; 140(2): 119-35, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23542924

ABSTRACT

The motor protein, prestin, situated in the basolateral plasma membrane of cochlear outer hair cells (OHCs), underlies the generation of somatic, voltage-driven mechanical force, the basis for the exquisite sensitivity, frequency selectivity and dynamic range of mammalian hearing. The molecular and structural basis of the ontogenetic development of this electromechanical force has remained elusive. The present study demonstrates that this force is significantly reduced when the immature subcellular distribution of prestin found along the entire plasma membrane persists into maturity, as has been described in previous studies under hypothyroidism. This observation suggests that cochlear amplification is critically dependent on the surface expression and distribution of prestin. Searching for proteins involved in organizing the subcellular localization of prestin to the basolateral plasma membrane, we identified cochlear expression of a novel truncated prestin splice isoform named prestin 9b (Slc26A5d) that contains a putative PDZ domain-binding motif. Using prestin 9b as the bait in a yeast two-hybrid assay, we identified a calcium/calmodulin-dependent serine protein kinase (CASK) as an interaction partner of prestin. Co-immunoprecipitation assays showed that CASK and prestin 9b can interact with full-length prestin. CASK was co-localized with prestin in a membrane domain where prestin-expressing OHC membrane abuts prestin-free OHC membrane, but was absent from this area for thyroid hormone deficiency. These findings suggest that CASK and the truncated prestin splice isoform contribute to confinement of prestin to the basolateral region of the plasma membrane. By means of such an interaction, the basal junction region between the OHC and its Deiter's cell may contribute to efficient generation of somatic electromechanical force.


Subject(s)
Anion Transport Proteins/metabolism , Electricity , Guanylate Kinases/metabolism , Hair Cells, Auditory, Outer/physiology , Mechanical Phenomena , Vestibular Nucleus, Lateral/cytology , Vestibular Nucleus, Lateral/metabolism , Animals , Anion Transport Proteins/analysis , Anion Transport Proteins/genetics , Cells, Cultured , Female , Guanylate Kinases/analysis , Guanylate Kinases/genetics , HEK293 Cells , Hair Cells, Auditory, Outer/chemistry , Hair Cells, Auditory, Outer/cytology , Humans , Immunohistochemistry , Mice , Mice, Inbred Strains , Molecular Motor Proteins/analysis , Molecular Motor Proteins/genetics , Molecular Motor Proteins/metabolism , Rats , Rats, Wistar , Sulfate Transporters , Vestibular Nucleus, Lateral/chemistry
18.
PLoS One ; 8(3): e57247, 2013.
Article in English | MEDLINE | ID: mdl-23516401

ABSTRACT

Tinnitus is proposed to be caused by decreased central input from the cochlea, followed by increased spontaneous and evoked subcortical activity that is interpreted as compensation for increased responsiveness of central auditory circuits. We compared equally noise exposed rats separated into groups with and without tinnitus for differences in brain responsiveness relative to the degree of deafferentation in the periphery. We analyzed (1) the number of CtBP2/RIBEYE-positive particles in ribbon synapses of the inner hair cell (IHC) as a measure for deafferentation; (2) the fine structure of the amplitudes of auditory brainstem responses (ABR) reflecting differences in sound responses following decreased auditory nerve activity and (3) the expression of the activity-regulated gene Arc in the auditory cortex (AC) to identify long-lasting central activity following sensory deprivation. Following moderate trauma, 30% of animals exhibited tinnitus, similar to the tinnitus prevalence among hearing impaired humans. Although both tinnitus and no-tinnitus animals exhibited a reduced ABR wave I amplitude (generated by primary auditory nerve fibers), IHCs ribbon loss and high-frequency hearing impairment was more severe in tinnitus animals, associated with significantly reduced amplitudes of the more centrally generated wave IV and V and less intense staining of Arc mRNA and protein in the AC. The observed severe IHCs ribbon loss, the minimal restoration of ABR wave size, and reduced cortical Arc expression suggest that tinnitus is linked to a failure to adapt central circuits to reduced cochlear input.


Subject(s)
Adaptation, Physiological , Cochlea/physiopathology , Evoked Potentials, Auditory, Brain Stem , Noise/adverse effects , Tinnitus/etiology , Animals , Auditory Cortex/metabolism , Auditory Threshold , Behavior, Animal , Cochlea/metabolism , Cytoskeletal Proteins/metabolism , Female , Hair Cells, Auditory, Inner/metabolism , Hearing Loss, Noise-Induced/physiopathology , Immunohistochemistry , Nerve Tissue Proteins/metabolism , Rats , Tinnitus/metabolism
19.
Mol Neurobiol ; 47(1): 261-79, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23154938

ABSTRACT

Increasing evidence shows that hearing loss is a risk factor for tinnitus and hyperacusis. Although both often coincide, a causal relationship between tinnitus and hyperacusis has not been shown. Currently, tinnitus and hyperacusis are assumed to be caused by elevated responsiveness in subcortical circuits. We examined both the impact of different degrees of cochlear damage and the influence of stress priming on tinnitus induction. We used (1) a behavioral animal model for tinnitus designed to minimize stress, (2) ribbon synapses in inner hair cells (IHCs) as a measure for deafferentation, (3) the integrity of auditory brainstem responses (ABR) to detect differences in stimulus-evoked neuronal activity, (4) the expression of the activity-regulated cytoskeletal protein, Arc, to identify long-lasting changes in network activity within the basolateral amygdala (BLA), hippocampal CA1, and auditory cortex (AC), and (5) stress priming to investigate the influence of corticosteroid on trauma-induced brain responses. We observed that IHC ribbon loss (deafferentation) leads to tinnitus when ABR functions remain reduced and Arc is not mobilized in the hippocampal CA1 and AC. If, however, ABR waves are functionally restored and Arc is mobilized, tinnitus does not occur. Both central response patterns were found to be independent of a profound threshold loss and could be shifted by the corticosterone level at the time of trauma. We, therefore, discuss the findings in the context of a history of stress that can trigger either an adaptive or nonadaptive brain response following injury.


Subject(s)
Cytoskeletal Proteins/metabolism , Hair Cells, Auditory, Inner/pathology , Nerve Tissue Proteins/metabolism , Noise/adverse effects , Tinnitus/metabolism , Tinnitus/pathology , Acoustic Stimulation , Animals , Auditory Cortex/metabolism , Auditory Cortex/pathology , Auditory Cortex/physiopathology , Auditory Threshold , Cytoskeletal Proteins/genetics , Evoked Potentials, Auditory, Brain Stem , Female , Hair Cells, Auditory, Inner/metabolism , Hearing Loss/complications , Hearing Loss/metabolism , Hearing Loss/pathology , Hearing Loss/physiopathology , Models, Biological , Nerve Tissue Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Stress, Psychological/complications , Stress, Psychological/pathology , Stress, Psychological/physiopathology , Tinnitus/complications , Tinnitus/physiopathology
20.
FASEB J ; 26(9): 3834-43, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22691916

ABSTRACT

Large conductance, voltage- and Ca(2+)-activated K(+) (BK) channels in inner hair cells (IHCs) of the cochlea are essential for hearing. However, germline deletion of BKα, the pore-forming subunit KCNMA1 of the BK channel, surprisingly did not affect hearing thresholds in the first postnatal weeks, even though altered IHC membrane time constants, decreased IHC receptor potential alternating current/direct current ratio, and impaired spike timing of auditory fibers were reported in these mice. To investigate the role of IHC BK channels for central auditory processing, we generated a conditional mouse model with hair cell-specific deletion of BKα from postnatal day 10 onward. This had an unexpected effect on temporal coding in the central auditory system: neuronal single and multiunit responses in the inferior colliculus showed higher excitability and greater precision of temporal coding that may be linked to the improved discrimination of temporally modulated sounds observed in behavioral training. The higher precision of temporal coding, however, was restricted to slower modulations of sound and reduced stimulus-driven activity. This suggests a diminished dynamic range of stimulus coding that is expected to impair signal detection in noise. Thus, BK channels in IHCs are crucial for central coding of the temporal fine structure of sound and for detection of signals in a noisy environment.


Subject(s)
Brain/physiology , Cochlea/physiology , Hair Cells, Auditory/physiology , Hearing/physiology , Large-Conductance Calcium-Activated Potassium Channels/physiology , Animals , Immunohistochemistry , Large-Conductance Calcium-Activated Potassium Channels/genetics , Learning , Mice , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...