Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
Front Mol Neurosci ; 6: 20, 2013.
Article in English | MEDLINE | ID: mdl-23950737

ABSTRACT

Voltage-gated L-type Ca(2+) channels (L-VGCCs) like CaV1.2 are assumed to play a crucial role for controlling release of trophic peptides including brain-derived neurotrophic factor (BDNF). In the inner ear of the adult mouse, besides the well-described L-VGCC CaV1.3, CaV1.2 is also expressed. Due to lethality of constitutive CaV1.2 knock-out mice, the function of this ion channel as well as its putative relationship to BDNF in the auditory system is entirely elusive. We recently described that BDNF plays a differential role for inner hair cell (IHC) vesicles release in normal and traumatized condition. To elucidate a presumptive role of CaV1.2 during this process, two tissue-specific conditional mouse lines were generated. To distinguish the impact of CaV1.2 on the cochlea from that on feedback loops from higher auditory centers CaV1.2 was deleted, in one mouse line, under the Pax2 promoter (CaV1.2(Pax2)) leading to a deletion in the spiral ganglion neurons, dorsal cochlear nucleus, and inferior colliculus. In the second mouse line, the Egr2 promoter was used for deleting CaV1.2 (CaV1.2(Egr2)) in auditory brainstem nuclei. In both mouse lines, normal hearing threshold and equal number of IHC release sites were observed. We found a slight reduction of auditory brainstem response wave I amplitudes in the CaV1.2(Pax2) mice, but not in the CaV1.2(Egr2) mice. After noise exposure, CaV1.2(Pax2) mice had less-pronounced hearing loss that correlated with maintenance of ribbons in IHCs and less reduced activity in auditory nerve fibers, as well as in higher brain centers at supra-threshold sound stimulation. As reduced cochlear BDNF mRNA levels were found in CaV1.2(Pax2) mice, we suggest that a CaV1.2-dependent step may participate in triggering part of the beneficial and deteriorating effects of cochlear BDNF in intact systems and during noise exposure through a pathway that is independent of CaV1.2 function in efferent circuits.

2.
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
3.
J Neurosci ; 32(25): 8545-53, 2012 Jun 20.
Article in English | MEDLINE | ID: mdl-22723694

ABSTRACT

The precision of sound information transmitted to the brain depends on the transfer characteristics of the inner hair cell (IHC) ribbon synapse and its multiple contacting auditory fibers. We found that brain derived neurotrophic factor (BDNF) differentially influences IHC characteristics in the intact and injured cochlea. Using conditional knock-out mice (BDNF(Pax2) KO) we found that resting membrane potentials, membrane capacitance and resting linear leak conductance of adult BDNF(Pax2) KO IHCs showed a normal maturation. Likewise, in BDNF(Pax2) KO membrane capacitance (ΔC(m)) as a function of inward calcium current (I(Ca)) follows the linear relationship typical for normal adult IHCs. In contrast the maximal ΔC(m), but not the maximal size of the calcium current, was significantly reduced by 45% in basal but not in apical cochlear turns in BDNF(Pax2) KO IHCs. Maximal ΔC(m) correlated with a loss of IHC ribbons in these cochlear turns and a reduced activity of the auditory nerve (auditory brainstem response wave I). Remarkably, a noise-induced loss of IHC ribbons, followed by reduced activity of the auditory nerve and reduced centrally generated wave II and III observed in control mice, was prevented in equally noise-exposed BDNF(Pax2) KO mice. Data suggest that BDNF expressed in the cochlea is essential for maintenance of adult IHC transmitter release sites and that BDNF upholds opposing afferents in high-frequency turns and scales them down following noise exposure.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Hair Cells, Auditory, Inner/physiology , Hearing Loss, Noise-Induced/genetics , Synapses/physiology , Animals , Blotting, Northern , Blotting, Western , Brain-Derived Neurotrophic Factor/genetics , Cell Count , Cochlea/growth & development , Cochlea/physiology , Evoked Potentials, Auditory, Brain Stem/physiology , Exocytosis/genetics , Exocytosis/physiology , Immunohistochemistry , Mice , Mice, Knockout , Noise/adverse effects , Otoacoustic Emissions, Spontaneous , PAX2 Transcription Factor/genetics , beta-Galactosidase/metabolism
4.
Mol Pharmacol ; 74(3): 595-604, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18524887

ABSTRACT

Tinnitus is a phantom auditory perception, which can be induced via application of concentrated sodium salicylate, and is known to be associated with hearing loss and altered neuronal excitability in peripheral and central auditory neurons. The molecular features of this excitability, however, has been poorly characterized to date. Brain-derived neurotrophic factor (BDNF), the activity-dependent cytoskeletal protein (Arg3.1, also known as Arc), and c-Fos are known to be affected by changes in excitability and plasticity. Using reverse transcription-polymerase chain reaction, in situ hybridization, and immunohistochemistry, the expression of these genes was monitored in the rat auditory system after local (cochlear) and systemic application of salicylate. Induction of tinnitus and hearing loss was verified in a behavioral model. Regardless of the mode of salicylate application, a common pattern became evident: 1) BDNF mRNA expression was increased in the spiral ganglion neurons of the cochlea; and 2) Arg3.1 expression was significantly reduced in the auditory cortex. Local application of the GABA(A) receptor modulator midazolam resulted in the reversal not only of salicylate-induced changes in cochlear BDNF expression, but also in cortical Arg3.1 expression, indicating that the tinnitus-associated changes in cochlear BDNF expression trigger the decline of cortical Arg3.1 expression. Furthermore, local midazolam application reduced tinnitus perception in the animal model. These findings support Arg3.1 and BDNF as markers for activity changes in the auditory system and suggest a role of GABAergic inhibition of cochlear neurons in the modulation of Arg3.1 plasticity changes in the auditory cortex and tinnitus perception.


Subject(s)
Auditory Perception/drug effects , Brain-Derived Neurotrophic Factor/genetics , Cytoskeletal Proteins/genetics , Gene Expression Regulation/drug effects , Midazolam/pharmacology , Nerve Tissue Proteins/genetics , Salicylates/pharmacology , Tinnitus/metabolism , Animals , Auditory Cortex/drug effects , Auditory Cortex/metabolism , Auditory Pathways/drug effects , Auditory Pathways/metabolism , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Cochlea/drug effects , Cochlea/metabolism , Cytoskeletal Proteins/metabolism , Female , Hearing Loss/chemically induced , Midazolam/administration & dosage , Models, Biological , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, GABA/metabolism , Salicylates/administration & dosage , Tinnitus/pathology
5.
Mol Pharmacol ; 73(4): 1085-91, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18198284

ABSTRACT

Brain-derived neurotrophic factor (BDNF) is a key neurotrophin whose expression is altered in response to neurological activity, influencing both short- and long-term synaptic changes. The BDNF gene consists of eight upstream exons (I-VII), each of which has a distinct promoter and can be independently spliced to the ninth coding exon (IX). We showed recently that the expression of BDNF exon IV in the cochlea is altered after exposure to salicylate, an ototoxic drug that in high doses is able to induce hearing loss and tinnitus. These changes were a crucial trigger for plasticity changes in the central auditory system. BDNF exon IV expression is regulated via interaction between calcium-response elements CaRE1, CaRE2, and CaRE3/Cre (CaREs) that are bound by the transcription factors CaRF1, upstream stimulatory factors 1 and 2 (USF1/2), and cAMP/Ca(2+) response element-binding protein (CREB), respectively. To determine whether the salicylate-induced changes in cochlear BDNF exon IV expression include a differential use of the CaRE binding proteins, we studied the level of the corresponding binding proteins in the spiral ganglion neurons before and after systemic application of concentrated salicylate using in situ hybridization and RT-PCR. BDNF exon IV and CaRF1 expression were up-regulated after application of salicylate, whereas USF1/2 and CREB mRNA expression remained unaffected. The changes in BDNF exon IV and CaRF1 expression were also dose-dependent. The data show Ca(2+) and CaRF1 as messengers of trauma (salicylate)-induced altered BDNF levels in the cochlea. Furthermore, they also provide the first evidence that a differential regulation of BDNF transcription factors might participate in BDNF-mediated plasticity changes.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cochlea/drug effects , Cochlea/metabolism , Gene Expression Regulation/drug effects , Salicylates/pharmacology , Transcription Factors/genetics , Transcription, Genetic/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Brain-Derived Neurotrophic Factor/genetics , Cochlea/cytology , Cyclic AMP Response Element-Binding Protein/metabolism , Dose-Response Relationship, Drug , Exons/genetics , Female , In Situ Hybridization , Injections , Neurons/drug effects , Neurons/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Rats , Rats, Wistar , Response Elements , Transcription Factors/metabolism
6.
Neurobiol Aging ; 28(4): 586-601, 2007 Apr.
Article in English | MEDLINE | ID: mdl-16580094

ABSTRACT

A decline in neuronal plasticity during the adult life span has been proposed to be associated with a reduced level of the effectors of plasticity responses (e.g., BDNF). Alteration of plasticity is also correlated with age-related hearing loss (presbycusis), but to date no detailed studies of BDNF expression have been performed in the young or aging mature cochlea. We have used rat and gerbil animal models for presbycusis, which displayed hearing loss in the final third of the animals' natural life span. We demonstrate for the first time a co-localization of BDNF protein, transcripts III and IV in cochlear neurons with a declining distribution towards low-frequency processing cochlear turns. BDNF protein was also found within the neuronal projections of the cochlea. A significant reduction of BDNF transcripts in high-frequency processing cochlear neurons was observed during aging, though this did not coincide with a major reduction of BDNF protein. In contrast, BDNF protein in peripheral and central projections was drastically reduced. Our results suggest that reduced BDNF protein levels in auditory nerves over age may be a crucial factor in the altered brainstem plasticity observed during presbycusis.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Gene Expression Regulation/physiology , Presbycusis/metabolism , RNA, Messenger/metabolism , Age Factors , Animals , Cell Count/methods , Cochlea/pathology , Disease Models, Animal , Evoked Potentials, Auditory, Brain Stem/physiology , Fluorescent Antibody Technique/methods , Gerbillinae , In Situ Hybridization/methods , Mice , Mice, Knockout , Neurons/metabolism , Presbycusis/genetics , Presbycusis/pathology , Presbycusis/physiopathology , Rats , Rats, Inbred F344 , Receptor, trkB/deficiency , Reverse Transcriptase Polymerase Chain Reaction/methods , Spiral Ganglion/pathology
7.
J Cell Sci ; 119(Pt 14): 2975-84, 2006 Jul 15.
Article in English | MEDLINE | ID: mdl-16803873

ABSTRACT

Thyroid hormone (TH or T3) and TH-receptor beta (TRbeta) have been reported to be relevant for cochlear development and hearing function. Mutations in the TRbeta gene result in deafness associated with resistance to TH syndrome. The effect of TRalpha1 on neither hearing function nor cochlear T3 target genes has been described to date. It is also uncertain whether TRalpha1 and TRbeta can act simultaneously on different target genes within a single cell. We focused on two concomitantly expressed outer hair cell genes, the potassium channel Kcnq4 and the motor protein prestin Slc26a5. In outer hair cells, TH enhanced the expression of the prestin gene through TRbeta. Simultaneously Kcnq4 expression was activated in the same cells by derepression of TRalpha1 aporeceptors mediated by an identified THresponse element, which modulates KCNQ4 promoter activity. We show that T3 target genes can differ in their sensitivity to TH receptors having the ligand either bound (holoreceptors) or not bound (aporeceptors) within single cells, and suggest a role for TRalpha1 in final cell differentiation.


Subject(s)
Cell Differentiation , Gene Expression Regulation , Hair Cells, Auditory, Outer/cytology , KCNQ Potassium Channels/genetics , Proteins/genetics , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormone Receptors beta/metabolism , Animals , Anion Transport Proteins , Base Sequence , Cells, Cultured , Genes, Dominant/genetics , Hair Cells, Auditory, Outer/metabolism , Humans , Hypothyroidism/metabolism , Mice , Molecular Sequence Data , Mutation/genetics , Promoter Regions, Genetic/genetics , Rats , Rats, Wistar , Response Elements/genetics , Sulfate Transporters , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors beta/genetics , Thyroid Hormones/deficiency
8.
Proc Natl Acad Sci U S A ; 101(35): 12922-7, 2004 Aug 31.
Article in English | MEDLINE | ID: mdl-15328414

ABSTRACT

The large conductance voltage- and Ca2+-activated potassium (BK) channel has been suggested to play an important role in the signal transduction process of cochlear inner hair cells. BK channels have been shown to be composed of the pore-forming alpha-subunit coexpressed with the auxiliary beta1-subunit. Analyzing the hearing function and cochlear phenotype of BK channel alpha-(BKalpha-/-) and beta1-subunit (BKbeta1-/-) knockout mice, we demonstrate normal hearing function and cochlear structure of BKbeta1-/- mice. During the first 4 postnatal weeks also, BKalpha-/- mice most surprisingly did not show any obvious hearing deficits. High-frequency hearing loss developed in BKalpha-/- mice only from approximately 8 weeks postnatally onward and was accompanied by a lack of distortion product otoacoustic emissions, suggesting outer hair cell (OHC) dysfunction. Hearing loss was linked to a loss of the KCNQ4 potassium channel in membranes of OHCs in the basal and midbasal cochlear turn, preceding hair cell degeneration and leading to a similar phenotype as elicited by pharmacologic blockade of KCNQ4 channels. Although the actual link between BK gene deletion, loss of KCNQ4 in OHCs, and OHC degeneration requires further investigation, data already suggest human BK-coding slo1 gene mutation as a susceptibility factor for progressive deafness, similar to KCNQ4 potassium channel mutations.


Subject(s)
Hearing Loss/genetics , Potassium Channels/genetics , Animals , Calcium/metabolism , Cochlea/metabolism , Gene Deletion , Hair Cells, Auditory, Outer/abnormalities , Hearing Loss/metabolism , Immunohistochemistry , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits , Mice , Phenotype , Potassium Channels/metabolism
9.
Development ; 130(19): 4741-50, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12925599

ABSTRACT

Members of the neurotrophin gene family and their high-affinity Trk receptors control innervation of the cochlea during embryonic development. Lack of neurotrophin signalling in the cochlea has been well documented for early postnatal animals, resulting in a loss of cochlear sensory neurones and a region-specific reduction of target innervation along the tonotopic axis. However, how reduced neurotrophin signalling affects the innervation of the mature cochlea is currently unknown. Here, we have analysed the consequences of a lack of the TrkB receptor and its ligand, the neurotrophin brain-derived neurotrophic factor (Bdnf), in the late postnatal or adult cochlea using mouse mutants. During early postnatal development, mutant animals show a lack of afferent innervation of outer hair cells in the apical part of the cochlea, whereas nerve fibres in the basal part are maintained. Strikingly, this phenotype is reversed during subsequent maturation of the cochlea, which results in a normal pattern of outer hair cell innervation in the apex and loss of nerve fibres at the base in adult mutants. Measurements of auditory brain stem responses of these mice revealed a significant hearing loss. The observed innervation patterns correlate with opposing gradients of Bdnf and Nt3 expression in cochlear neurones along the tonotopic axis. Thus, the reshaping of innervation may be controlled by autocrine signalling between neurotrophins and their receptors in cochlear neurones. Our results indicate a substantial potential for re-innervation processes in the mature cochlea, which may also be of relevance for treatment of hearing loss in humans.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cochlea/growth & development , Cochlea/innervation , Hearing Loss, Sensorineural/etiology , Membrane Glycoproteins , Receptor, trkB/metabolism , Signal Transduction/physiology , Animals , Anion Transport Proteins , Brain-Derived Neurotrophic Factor/genetics , Cochlea/anatomy & histology , Cochlea/metabolism , Evoked Potentials, Auditory , Humans , Intermediate Filament Proteins/metabolism , Mice , Molecular Motor Proteins , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Neurotrophin 3/genetics , Neurotrophin 3/metabolism , Peripherins , Proteins/metabolism , Receptor, trkB/genetics , Sulfate Transporters , Synapses/metabolism
10.
Proc Natl Acad Sci U S A ; 99(5): 2901-6, 2002 Mar 05.
Article in English | MEDLINE | ID: mdl-11867734

ABSTRACT

The most impressive property of outer hair cells (OHCs) is their ability to change their length at high acoustic frequencies, thus providing the exquisite sensitivity and frequency-resolving capacity of the mammalian hearing organ. Prestin, a protein related to a sulfate/anion transport protein, recently has been identified and proposed as the OHC motor molecule. Homology searches of 1.5 kb of genomic DNA 5' of the coding region of the prestin gene allowed the identification of a thyroid hormone (TH) response element (TRE) in the first intron upstream of the prestin ATG codon. Prestin(TRE) bound TH receptors as a monomer or presumptive heterodimer and mediated a triiodothyronine-dependent transactivation of a heterologous promotor in response to triiodothyronine receptors alpha and beta. Retinoid X receptor-alpha had an additive effect. Expression of prestin mRNA and prestin protein was reduced strongly in the absence of TH. Although prestin protein typically was redistributed to the lateral membrane before the onset of hearing, an immature pattern of prestin protein distribution across the entire OHC membrane was noted in hypothyroid rats. The data suggest TH as a first transcriptional regulator of the motor protein prestin and as a direct or indirect modulator of subcellular prestin distribution.


Subject(s)
Proteins/genetics , Receptors, Thyroid Hormone , Trans-Activators/metabolism , Transcriptional Activation , Triiodothyronine/metabolism , 5' Flanking Region , Animals , Anion Transport Proteins , Base Sequence , Cell Line , Cochlea/metabolism , DNA, Complementary , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genes, Reporter , Hair Cells, Auditory, Outer/metabolism , HeLa Cells , Humans , Luciferases/genetics , Molecular Sequence Data , Proteins/metabolism , Rats , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Response Elements , Sulfate Transporters , Trans-Activators/pharmacology , Transfection , Triiodothyronine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL