Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(4)2023 Feb 07.
Article in English | MEDLINE | ID: mdl-36834731

ABSTRACT

Endogenous glucocorticoids (GC) are known to modulate basic elements of cochlear physiology. These include both noise-induced injury and circadian rhythms. While GC signaling in the cochlea can directly influence auditory transduction via actions on hair cells and spiral ganglion neurons, evidence also indicates that GC signaling exerts effects via tissue homeostatic processes that can include effects on cochlear immunomodulation. GCs act at both the glucocorticoid receptor (GR) and the mineralocorticoid receptor (MR). Most cell types in the cochlea express both receptors sensitive to GCs. The GR is associated with acquired sensorineural hearing loss (SNHL) through its effects on both gene expression and immunomodulatory programs. The MR has been associated with age-related hearing loss through dysfunction of ionic homeostatic balance. Cochlear supporting cells maintain local homeostatic requirements, are sensitive to perturbation, and participate in inflammatory signaling. Here, we have used conditional gene manipulation techniques to target Nr3c1 (GR) or Nr3c2 (MR) for tamoxifen-induced gene ablation in Sox9-expressing cochlear supporting cells of adult mice to investigate whether either of the receptors sensitive to GCs plays a role in protecting against (or exacerbating) noise-induced cochlear damage. We have selected mild intensity noise exposure to examine the role of these receptors related to more commonly experienced noise levels. Our results reveal distinct roles of these GC receptors for both basal auditory thresholds prior to noise exposure and during recovery from mild noise exposure. Prior to noise exposure, auditory brainstem responses (ABRs) were measured in mice carrying the floxed allele of interest and the Cre recombinase transgene, but not receiving tamoxifen injections (defined as control (no tamoxifen treatment), versus conditional knockout (cKO) mice, defined as mice having received tamoxifen injections. Results revealed hypersensitive thresholds to mid- to low-frequencies after tamoxifen-induced GR ablation from Sox9-expressing cochlear supporting cells compared to control (no tamoxifen) mice. GR ablation from Sox9-expressing cochlear supporting cells resulted in a permanent threshold shift in mid-basal cochlear frequency regions after mild noise exposure that produced only a temporary threshold shift in both control (no tamoxifen) f/fGR:Sox9iCre+ and heterozygous f/+GR:Sox9iCre+ tamoxifen-treated mice. A similar comparison of basal ABRs measured in control (no tamoxifen) and tamoxifen-treated, floxed MR mice prior to noise exposure indicated no difference in baseline thresholds. After mild noise exposure, MR ablation was initially associated with a complete threshold recovery at 22.6 kHz by 3 days post-noise. Threshold continued to shift to higher sensitivity over time such that by 30 days post-noise exposure the 22.6 kHz ABR threshold was 10 dB more sensitive than baseline. Further, MR ablation produced a temporary reduction in peak 1 neural amplitude one day post-noise. While supporting cell GR ablation trended towards reducing numbers of ribbon synapses, MR ablation reduced ribbon synapse counts but did not exacerbate noise-induced damage including synapse loss at the experimental endpoint. GR ablation from the targeted supporting cells increased the basal resting number of Iba1-positive (innate) immune cells (no noise exposure) and decreased the number of Iba1-positive cells seven days following noise exposure. MR ablation did not alter innate immune cell numbers at seven days post-noise exposure. Taken together, these findings support differential roles of cochlear supporting cell MR and GR expression at basal, resting conditions and especially during recovery from noise exposure.


Subject(s)
Hearing Loss, Noise-Induced , Mice , Animals , Hearing Loss, Noise-Induced/metabolism , Glucocorticoids/metabolism , Receptors, Mineralocorticoid/metabolism , Cochlea/metabolism , Hearing , Auditory Threshold/physiology , Receptors, Glucocorticoid/metabolism
2.
Methods Mol Biol ; 2585: 51-69, 2023.
Article in English | MEDLINE | ID: mdl-36331765

ABSTRACT

Immunohistochemistry is a valuable tool for probing not only scientific questions but also clinical diagnoses. It provides power from localization of a protein within the milieu of a tissue section that may reflect positioning within or beyond the boundaries of a cell that is representative of the tissue at a discrete moment in time. The method can be applied broadly, including to tissues under normal, developmental, chemically, or genetically altered conditions and disease states.Disease manifesting from West Nile virus infection ranges from acute, systemic febrile symptoms to compromise of central nervous system function. Immunohistochemistry has been used to assess WNV infection in the nervous system in postmortem and experimental conditions, despite the lack of understanding of the precise route of viral entry. In addition to imprecise knowledge of initial viral entry into cells and whether entry is even the same between cell types, the fact that spontaneous viral mutations and environmental pressures from climate change may alter the prevalence of the disease state across geographical and climatological boundaries highlights the need for continued assessment of infection. Immunohistochemistry is a useful way to assess these aspects of WNV infection with the aim being to better understand the organs and cell types that are compromised by WNV infection. This chapter outlines how this can be carried out on brain tissue, but the procedures discussed can also be applied more broadly on tissue outside of the central nervous system.


Subject(s)
West Nile Fever , West Nile virus , Humans , West Nile Fever/diagnosis , Viral Envelope Proteins , Brain , Immunohistochemistry
3.
Otol Neurotol ; 42(3): 466-474, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33351563

ABSTRACT

HYPOTHESIS: Localized cooling of the external ear has a protective effect on the susceptibility to cisplatin-induced hearing loss. BACKGROUND: We previously demonstrated significant protection from cisplatin-induced hearing loss using cool water ear canal irrigation. However, the study was limited to a single bolus injection of cisplatin and an acute time period. Here, we examined the application of localized cooling of the ear canal with repeated doses of cisplatin, over an expanded period of time, and using two methods of cooling. METHODS: Twenty-four guinea pigs (12 male and 12 female) underwent auditory physiological testing (auditory brainstem response and distortion product otoacoustic emissions at 8-32 kHz) and pre/postadministration of cisplatin. Cisplatin (4 mg/kg i.p.) was administered in 3 weekly single injections for a total of 12 mg/kg. While anesthetized, the left ears of the guinea pigs were exposed to either cool water (22°C; ICS Water Caloric Irrigator), a cool ear bar (15°C, cooled by a Peltier device; TNM, Scion NeuroStim), or left uncooled as a sham control. The animals were tested 3 days post each dosage and 1 month post the final dose. At the end of the experiment the animals were euthanized for histological evaluation. RESULTS: We found that hearing loss was significantly reduced, and hair cell survival greatly improved, in animals that received cooling treatments compared to cisplatin-only control animals. No significant difference was observed between the two methods of cooling. CONCLUSION: Localized cooling of the ear canal during administration of cisplatin mitigated loss of auditory function and loss of hair cells.


Subject(s)
Antineoplastic Agents , Hearing Loss , Animals , Antineoplastic Agents/adverse effects , Cisplatin/toxicity , Evoked Potentials, Auditory, Brain Stem , Female , Guinea Pigs , Hair Cells, Auditory , Hearing , Hearing Loss/chemically induced , Hearing Loss/drug therapy , Hearing Loss/prevention & control , Male , Otoacoustic Emissions, Spontaneous
4.
Hear Res ; 395: 108000, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32623238

ABSTRACT

Zika virus (ZIKV) has been recently recognized as a causative agent of newborn microcephaly, as well as other neurological consequences. A less well recognized comorbidity of prenatal ZIKV infection is hearing loss, but cases of hearing impairment following adult ZIKV infection have also been recognized. Diminished hearing following prenatal ZIKV infection in a mouse model has been reported, but no cellular consequences were observed. We examined the effects of ZIKV infection on inner ear cellular integrity and expression levels of various proteins important for cochlear function in type I interferon receptor null (Ifnar1-/-) mice following infection at 5-6 weeks of age. We show that ZIKV antigens are present in cells within the cochlear epithelium, lateral wall, spiral limbus and spiral ganglion. Here we show that ZIKV infection alters cochlear expression of genes that signal cell damage (S100B), transport fluids (AQP1), are gaseous transmitters (eNOs) and modulate immune response (F4/80). Morphological analyses shows that not only are cochlear structures compromised by ZIKV infection, but damage also occurs in vestibular end organs. ZIKV produces a graded distribution of cellular damage in the cochlea, with greatest damage in the apex similar to that reported for cytomegalovirus (CMV) infection. The graded distribution of damage may indicate a differential susceptibility to ZIKV along the cochlear tonotopic axis. Collectively, these data are the first to show the molecular and morphological damage to the inner ear induced by ZIKV infection in adults and suggests multiple mechanisms contributing to the hearing loss reported in the human population.


Subject(s)
Ear, Inner , Zika Virus Infection , Zika Virus , Animals , Cytomegalovirus Infections , Disease Models, Animal , Female , Hearing Loss , Mice , Pregnancy , Zika Virus Infection/complications
5.
Hear Res ; 388: 107896, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31982642

ABSTRACT

The development of knockin mice with Cre recombinase expressed under the control of the promoter for choline acetyltransferase (ChAT) has allowed experimental manipulation of cholinergic circuits. However, currently available ChATCre mouse lines are on the C57BL/6J strain background, which shows early onset age-related hearing loss attributed to the Cdh23753A mutation (a.k.a., the ahl mutation). To develop ChATCre mice without accelerated hearing loss, we backcrossed ChATIRES-Cre mice with CBA/CaJ mice that have normal hearing. We used genotyping to obtain mice homozygous for ChATIRES-Cre and the wild-type allele at the Cdh23 locus (ChATCre,Cdh23WT). In the new line, auditory brainstem response thresholds were ∼20 dB lower than those in 9 month old ChATIRES-Cre mice at all frequencies tested (4-31.5 kHz). These thresholds were stable throughout the period of testing (3-12 months of age). We then bred ChATCre,Cdh23WT animals with Ai14 reporter mice to confirm the expression pattern of ChATCre. In these mice, tdTomato-labeled cells were observed in all brainstem regions known to contain cholinergic cells. We then stained the tissue with a neuron-specific marker, NeuN, to determine whether Cre expression was limited to neurons. Across several brainstem nuclei (pontomesencephalic tegmentum, motor trigeminal and facial nuclei), 100% of the tdTomato-labeled cells were double-labeled with anti-NeuN (n = 1896 cells), indicating Cre-recombinase was limited to neurons. Almost all of these cells (1867/1896 = 98.5%) also stained with antibodies against ChAT, indicating that reporter label was expressed almost exclusively in cholinergic neurons. Finally, an average 88.7% of the ChAT+ cells in these nuclei were labeled with tdTomato, indicating that the Cre is expressed in a large proportion of the cholinergic cells in these nuclei. We conclude that the backcrossed ChATCre,Cdh23WT mouse line has normal hearing and expresses Cre recombinase almost exclusively in cholinergic neurons. This ChATCre,Cdh23WT mouse line may provide an opportunity to manipulate cholinergic circuits without the confound of accelerated hearing loss associated with the C57BL/6J background. Furthermore, comparison with lines that do show early hearing loss may provide insight into possible cholinergic roles in age-related hearing loss.


Subject(s)
Brain Stem/enzymology , Choline O-Acetyltransferase/metabolism , Cholinergic Fibers/enzymology , Hearing Loss/prevention & control , Hearing , Integrases/metabolism , Animals , Auditory Threshold , Brain Stem/physiopathology , Cadherins/genetics , Choline O-Acetyltransferase/genetics , Crosses, Genetic , DNA-Binding Proteins/metabolism , Evoked Potentials, Auditory, Brain Stem , Female , Gene Knock-In Techniques , Hearing Loss/enzymology , Hearing Loss/genetics , Hearing Loss/physiopathology , Integrases/genetics , Male , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Mutation , Nerve Tissue Proteins/metabolism , Promoter Regions, Genetic , Species Specificity
6.
Bone ; 120: 285-296, 2019 03.
Article in English | MEDLINE | ID: mdl-30414510

ABSTRACT

Alterations in bone strength and structure were found in knockout (KO) mouse strains with deletion of several acetylcholine receptors. Interestingly, the expression of the nicotinic acetylcholine receptors (nAChR) subunit α10 was down-regulated in osteogenic differentiated mesenchymal stem cells of patients with osteoporosis whereas the expression of subunit α9 was not altered. Since nAChR subunits α9 and α10 are often combined in a functional receptor, we analyzed here the bone of adult female KO mice with single deletion of either nAChR alpha9 (α9KO) or alpha10 (α10KO). Biomechanical testing showed a significant decrease of bending stiffness and maximal breaking force in α9KO compared to their corresponding wild type mice. Furthermore, an increase in trabecular pattern factor (Tb.Pf) and structure model index (SMI) was detected by µCT in α9KO indicating reduced bone mass. On the mRNA level a decrease of Collagen 1α1 and Connexin-43 was measured by real-time RT-PCR in α9KO while no alteration of osteoclast markers was detected in either mouse strain. Using electron microcopy we observed an increase in the number of osteocytes that showed signs of degeneration and cell death in the α9KO compared to their wild type mice, while α10KO showed no differences. In conclusion, we demonstrate alterations in bone strength, structure and bio-marker expression in α9KO mice which imply the induction of osteocyte degeneration. Thus, our data suggest that nAChR containing the α9 subunit might be involved in the homeostasis of osteocytes and therefore in bone mass regulation.


Subject(s)
Bone and Bones/anatomy & histology , Gene Deletion , Receptors, Nicotinic/genetics , Alkaline Phosphatase/metabolism , Animals , Animals, Newborn , Bone and Bones/physiology , Cancellous Bone/anatomy & histology , Cortical Bone/anatomy & histology , Female , Femur/anatomy & histology , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteocytes/metabolism , Osteocytes/ultrastructure , Receptors, Nicotinic/deficiency
7.
Front Cell Neurosci ; 11: 282, 2017.
Article in English | MEDLINE | ID: mdl-28955208

ABSTRACT

The α9 and α10 nicotinic acetylcholine receptor (nAChR) subunits are likely to be the evolutionary precursors to the entire cys-loop superfamily of ligand-gated ion channels, which includes acetylcholine, GABA, glycine and serotonin ionotropic receptors. nAChRs containing α9 and α10 subunits are found in the inner ear, dorsal root ganglia and many non-excitable tissues, but their expression in the central nervous system has not been definitely demonstrated. Here we show the presence of both α9 and α10 nAChR subunits in the mouse brain by RT-PCR and immunochemical approaches with a range of nAChR subunit-selective antibodies, which selectivity was demonstrated in the brain preparations of α7-/-, α9-/- and α10-/- mice. The α9 and α10 RNA transcripts were found in medulla oblongata (MO), cerebellum, midbrain (MB), thalamus and putamen (TP), somatosensory cortex (SC), frontal cortex (FC) and hippocampus. High α9-selective signal in ELISA was observed in the FC, SC, MO, TP and hippocampus and α10-selective signal was the highest in MO and FC. The α9 and α10 proteins were found in the brain mitochondria, while their presence on the plasma membrane has not been definitely confirmed The α7-, α9- and α10-selective antibodies stained mainly neurons and hypertrophied astrocytes, but not microglia. The α9- and α10-positive cells formed ordered structures or zones in cerebellum and superior olive (SO) and were randomly distributed among α7-positive cells in the FC; they were found in CA1, CA3 and CA4, but not in CA2 region of the hippocampus. The α9 and α10 subunits were up-regulated in α7-/- mice and both α7 and α9 subunits were down-regulated in α10-/- mice. We conclude that α9 and α10 nAChR subunits are expressed in distinct neurons of the mouse brain and in the brain mitochondria and are compensatory up-regulated in the absence of α7 subunits.

8.
Proc Natl Acad Sci U S A ; 114(10): E1825-E1832, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28223528

ABSTRACT

Opioids are first-line drugs for moderate to severe acute pain and cancer pain. However, these medications are associated with severe side effects, and whether they are efficacious in treatment of chronic nonmalignant pain remains controversial. Medications that act through alternative molecular mechanisms are critically needed. Antagonists of α9α10 nicotinic acetylcholine receptors (nAChRs) have been proposed as an important nonopioid mechanism based on studies demonstrating prevention of neuropathology after trauma-induced nerve injury. However, the key α9α10 ligands characterized to date are at least two orders of magnitude less potent on human vs. rodent nAChRs, limiting their translational application. Furthermore, an alternative proposal that these ligands achieve their beneficial effects by acting as agonists of GABAB receptors has caused confusion over whether blockade of α9α10 nAChRs is the fundamental underlying mechanism. To address these issues definitively, we developed RgIA4, a peptide that exhibits high potency for both human and rodent α9α10 nAChRs, and was at least 1,000-fold more selective for α9α10 nAChRs vs. all other molecular targets tested, including opioid and GABAB receptors. A daily s.c. dose of RgIA4 prevented chemotherapy-induced neuropathic pain in rats. In wild-type mice, oxaliplatin treatment produced cold allodynia that could be prevented by RgIA4. Additionally, in α9 KO mice, chemotherapy-induced development of cold allodynia was attenuated and the milder, temporary cold allodynia was not relieved by RgIA4. These findings establish blockade of α9-containing nAChRs as the basis for the efficacy of RgIA4, and that α9-containing nAChRs are a critical target for prevention of chronic cancer chemotherapy-induced neuropathic pain.


Subject(s)
Cancer Pain/drug therapy , Hyperalgesia/drug therapy , Peptides/administration & dosage , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Analgesics, Opioid/adverse effects , Animals , Cancer Pain/chemically induced , Cancer Pain/genetics , Cancer Pain/pathology , Humans , Hyperalgesia/chemically induced , Hyperalgesia/genetics , Hyperalgesia/pathology , Ligands , Mice , Mice, Knockout , Neuralgia/chemically induced , Neuralgia/drug therapy , Neuralgia/genetics , Neuralgia/pathology , Nicotinic Antagonists/administration & dosage , Organoplatinum Compounds/adverse effects , Oxaliplatin , Receptors, GABA-B/genetics
9.
Methods Mol Biol ; 1427: 109-33, 2016.
Article in English | MEDLINE | ID: mdl-27259924

ABSTRACT

Modern biologists have at their disposal a large array of techniques used to assess the existence and relative or absolute quantity of any molecule of interest in a sample. However, implementing most of these procedures can be a daunting task for the first time, even in a lab with experienced researchers. Just choosing a protocol to follow can take weeks while all of the nuances are examined and it is determined whether a protocol will (a) give the desired results, (b) result in interpretable and unbiased data, and (c) be amenable to the sample of interest. We detail here a robust procedure for labeling proteins in a complex lysate for the ultimate differential quantification of protein abundance following experimental manipulations. Following a successful outcome of the labeling procedure, the sample is submitted for mass spectrometric analysis, resulting in peptide quantification and protein identification. While we will concentrate on cells in culture, we will point out procedures that can be used for labeling lysates generated from tissues, along with any minor modifications required for such samples. We will also outline, but not fully document, other strategies used in our lab to label proteins prior to mass spectrometric analysis, and describe under which conditions each procedure may be desirable. What is not covered in this chapter is anything but the most brief introduction to mass spectrometry (instrumentation, theory, etc.), nor do we attempt to cover much in the way of software used for post hoc analysis. These two topics are dependent upon one's resources, and where applicable, one's collaborators. We strongly encourage the reader to seek out expert advice on topics not covered here.


Subject(s)
Ear, Inner/metabolism , Isotope Labeling/methods , Proteomics/methods , Animals , Cells, Cultured , Cochlea/metabolism , Humans , Mass Spectrometry , Proteins/chemistry , Proteins/isolation & purification
10.
Biochem Pharmacol ; 97(1): 1-15, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26074267

ABSTRACT

Hearing loss afflicts approximately 15% of the world's population, and crosses all socioeconomic boundaries. While great strides have been made in understanding the genetic components of syndromic and non-syndromic hearing loss, understanding of the mechanisms underlying noise-induced hearing loss (NIHL) have come much more slowly. NIHL is not simply a mechanism by which older individuals loose their hearing. Significantly, the incidence of NIHL is increasing, and is now involving ever younger populations. This may predict future increased occurrences of hearing loss. Current research has shown that even short-term exposures to loud sounds generating what was previously considered temporary hearing loss, actually produces an almost immediate and permanent loss of specific populations of auditory nerve fibers. Additionally, recurrent exposures to intense sound may hasten age-related hearing loss. While NIHL is a significant medical concern, to date, few compounds have delivered significant protection, arguing that new targets need to be identified. In this commentary, we will explore cellular signaling processes taking place in the cochlea believed to be involved in protection against hearing loss, and highlight new data suggestive of novel signaling not previously recognized as occurring in the cochlea, that is perhaps protective of hearing. This includes a recently described local hypothalamic-pituitary-adrenal axis (HPA)-like signaling system fully contained in the cochlea. This system may represent a local cellular stress-response system based on stress hormone release similar to the systemic HPA axis. Its discovery may hold hope for new drug therapies that can be delivered directly to the cochlea, circumventing systemic side effects.


Subject(s)
Cochlea/metabolism , Corticotropin-Releasing Hormone/metabolism , Hearing Loss, Noise-Induced/metabolism , Models, Biological , Receptors, Corticotropin-Releasing Hormone/agonists , Signal Transduction , Stress, Physiological/radiation effects , Animals , Cochlea/innervation , Cochlea/radiation effects , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/radiation effects , Hearing Loss, Noise-Induced/prevention & control , Humans , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/radiation effects , Neurons/metabolism , Neurons/radiation effects , Noise/adverse effects , Oxidative Stress/radiation effects , Pituitary-Adrenal System/metabolism , Pituitary-Adrenal System/radiation effects , Receptors, Corticotropin-Releasing Hormone/metabolism , Signal Transduction/radiation effects , Superior Olivary Complex/metabolism , Superior Olivary Complex/radiation effects
11.
Cerebellum ; 14(5): 557-69, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25592068

ABSTRACT

Although the major emphasis of Enrico Mugnaini's research has been on investigations of the cerebellum, a significant amount of work over a relatively short span of time was also done in his lab on a number of other brain systems. These centered on sensory systems. One of these extra-cerebellar systems that he embraced was the auditory system. Portions of the cochlear nucleus, the first synaptic relay station along the central auditory pathways, possess a cerebellar-like circuitry and neurochemistry, and this no doubt lured Enrico into the auditory field. As new tools became available to pursue neuroanatomical research in general, which included a novel antibody to glutamic acid decarboxylase (GAD), Enrico's lab soon branched out into investigating many other brain structures beyond the cerebellum, with an overall goal of producing a map illustrating GAD expression in the brain. In collaboration with long-term colleagues, one of these many non-cerebellar regions he took an interest in was an efferent pathway originating in the superior olive and projecting to the cochlea, the peripheral end organ for hearing. There was a need for a more complete neurochemical map of this olivocochlear efferent system, and armed with new antibodies and well-established tract tracing tools, together we set out to further explore this system. This short review describes the work done with Enrico on the olivocochlear system of rodents, and also continues the story beyond Enrico's lab to reveal how the work done in his lab fits into the larger scheme of current, ongoing research into the olivocochlear system.


Subject(s)
Cerebellum/physiology , Mammals/anatomy & histology , Olivary Nucleus/anatomy & histology , Olivary Nucleus/physiology , Animals , Auditory Pathways/physiology , History, 20th Century , Humans , Neuroanatomy/history , Neurochemistry/history
12.
Neuron ; 82(4): 822-35, 2014 May 21.
Article in English | MEDLINE | ID: mdl-24853941

ABSTRACT

Patterned spontaneous activity is a hallmark of developing sensory systems. In the auditory system, rhythmic bursts of spontaneous activity are generated in cochlear hair cells and propagated along central auditory pathways. The role of these activity patterns in the development of central auditory circuits has remained speculative. Here we demonstrate that blocking efferent cholinergic neurotransmission to developing hair cells in mice that lack the α9 subunit of nicotinic acetylcholine receptors (α9 KO mice) altered the temporal fine structure of spontaneous activity without changing activity levels. KO mice showed a severe impairment in the functional and structural sharpening of an inhibitory tonotopic map, as evidenced by deficits in synaptic strengthening and silencing of connections and an absence in axonal pruning. These results provide evidence that the precise temporal pattern of spontaneous activity before hearing onset is crucial for the establishment of precise tonotopy, the major organizing principle of central auditory pathways.


Subject(s)
Action Potentials/physiology , Auditory Pathways/physiology , Brain Mapping , Brain Stem/cytology , Action Potentials/genetics , Age Factors , Animals , Animals, Newborn , Auditory Pathways/growth & development , Biophysics , Brain Stem/growth & development , Electric Stimulation , Functional Laterality/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Inhibition/genetics , Noise , Olivary Nucleus/cytology , Olivary Nucleus/growth & development , Receptors, Nicotinic/deficiency
13.
Open Biol ; 3(11): 130163, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24350389

ABSTRACT

Spontaneous electrical activity generated by developing sensory cells and neurons is crucial for the maturation of neural circuits. The full maturation of mammalian auditory inner hair cells (IHCs) depends on patterns of spontaneous action potentials during a 'critical period' of development. The intrinsic spiking activity of IHCs can be modulated by inhibitory input from cholinergic efferent fibres descending from the brainstem, which transiently innervate immature IHCs. However, it remains unknown whether this transient efferent input to developing IHCs is required for their functional maturation. We used a mouse model that lacks the α9-nicotinic acetylcholine receptor subunit (α9nAChR) in IHCs and another lacking synaptotagmin-2 in the efferent terminals to remove or reduce efferent input to IHCs, respectively. We found that the efferent system is required for the developmental linearization of the Ca(2+)-sensitivity of vesicle fusion at IHC ribbon synapses, without affecting their general cell development. This provides the first direct evidence that the efferent system, by modulating IHC electrical activity, is required for the maturation of the IHC synaptic machinery. The central control of sensory cell development is unique among sensory systems.


Subject(s)
Hair Cells, Auditory, Inner/physiology , Motor Neurons/physiology , Receptors, Nicotinic/physiology , Synapses/physiology , Synaptic Transmission/physiology , Action Potentials , Animals , Cochlea/physiology , Mice , Mice, Knockout , Receptors, Nicotinic/genetics , Stereocilia , Synaptotagmin II/genetics , Synaptotagmin II/physiology
14.
J Neurophysiol ; 108(2): 491-500, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22514298

ABSTRACT

Suppression of ipsilateral distortion product otoacoustic emissions (DPOAEs) by contralateral noise is used in humans and animals to assay the strength of sound-evoked negative feedback from the medial olivocochlear (MOC) efferent pathway. However, depending on species and anesthesia, contributions of other feedback systems to the middle or inner ear can cloud the interpretation. Here, contributions of MOC and middle-ear muscle reflexes, as well as autonomic feedback, to contra-noise suppression in anesthetized mice are dissected by selectively eliminating each pathway by surgical transection, pharmacological blockade, or targeted gene deletion. When ipsilateral DPOAEs were evoked by low-level primaries, contra-noise suppression was typically ~1 dB with contra-noise levels around 95 dB SPL, and it always disappeared upon contralateral cochlear destruction. Lack of middle-ear muscle contribution was suggested by persistence of contra-noise suppression after paralysis with curare, tensor tympani cauterization, or section of the facial nerve. Contribution of cochlear sympathetics was ruled out by studying mutant mice lacking adrenergic signaling (dopamine ß-hydroxylase knockouts). Surprisingly, contra-noise effects on low-level DPOAEs were also not diminished by eliminating the MOC system pharmacologically (strychnine), surgically, or by deletion of relevant cholinergic receptors (α9/α10). In contrast, when ipsilateral DPOAEs were evoked by high-level primaries, the contra-noise suppression, although comparable in magnitude, was largely eliminated by MOC blockade or section. Possible alternate pathways are discussed for the source of contra-noise-evoked effects at low ipsilateral levels.


Subject(s)
Anesthesia, General , Biofeedback, Psychology/physiology , Cochlea/physiology , Muscle, Skeletal/physiology , Olivary Nucleus/physiology , Otoacoustic Emissions, Spontaneous/physiology , Afferent Pathways/physiology , Animals , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal-To-Noise Ratio
15.
Hear Res ; 288(1-2): 3-18, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22484018

ABSTRACT

A key property possessed by the mammalian cochlea is its ability to dynamically alter its own sensitivity. Because hair cells and ganglion cells are prone to damage following exposure to loud sound, extant mechanisms limiting cochlear damage include modulation involving both the mechanical (via outer hair cell motility) and neural signaling (via inner hair cell-ganglion cell synapses) steps of peripheral auditory processing. Feedback systems such as that embodied by the olivocochlear system can alter sensitivity, but respond only after stimulus encoding, allowing potentially damaging sounds to impact the inner ear before sensitivity is adjusted. Less well characterized are potential cellular signaling systems involved in protection against metabolic stress and resultant damage. Although pharmacological manipulation of the olivocochlear system may hold some promise for attenuating cochlear damage, targeting this system may still allow damage to occur that does not depend on a fully functional feedback loop for its mitigation. Thus, understanding endogenous cell signaling systems involved in cochlear protection may lead to new strategies and therapies for prevention of cochlear damage and consequent hearing loss. We have recently discovered a novel cochlear signaling system that is molecularly equivalent to the classic hypothalamic-pituitary-adrenal (HPA) axis. This cochlear HPA-equivalent system functions to balance auditory sensitivity and susceptibility to noise-induced hearing loss, and also protects against cellular metabolic insults resulting from exposures to ototoxic drugs. This system may represent a local cellular response system designed to mitigate damage arising from various types of insult.


Subject(s)
Cochlea/metabolism , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Signal Transduction , Animals , Auditory Pathways/metabolism , Cochlea/drug effects , Cochlea/pathology , Feedback, Physiological , Glucocorticoids/metabolism , Hearing Loss, Noise-Induced/metabolism , Hearing Loss, Noise-Induced/pathology , Hearing Loss, Noise-Induced/prevention & control , Humans , Noise/adverse effects , Receptors, Corticotropin-Releasing Hormone/metabolism , Signal Transduction/drug effects
16.
Mol Neurobiol ; 44(3): 383-406, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21909974

ABSTRACT

A key requirement for encoding the auditory environment is the ability to dynamically alter cochlear sensitivity. However, merely attaining a steady state of maximal sensitivity is not a viable solution since the sensory cells and ganglion cells of the cochlea are prone to damage following exposure to loud sound. Most often, such damage is via initial metabolic insult that can lead to cellular death. Thus, establishing the highest sensitivity must be balanced with protection against cellular metabolic damage that can lead to loss of hair cells and ganglion cells, resulting in loss of frequency representation. While feedback mechanisms are known to exist in the cochlea that alter sensitivity, they respond only after stimulus encoding, allowing potentially damaging sounds to impact the inner ear at times coincident with increased sensitivity. Thus, questions remain concerning the endogenous signaling systems involved in dynamic modulation of cochlear sensitivity and protection against metabolic stress. Understanding endogenous signaling systems involved in cochlear protection may lead to new strategies and therapies for prevention of cochlear damage and consequent hearing loss. We have recently discovered a novel cochlear signaling system that is molecularly equivalent to the classic hypothalamic-pituitary-adrenal (HPA) axis. This cochlear HPA-equivalent system functions to balance auditory sensitivity and susceptibility to noise-induced hearing loss, and also protects against cellular metabolic insults resulting from exposures to ototoxic drugs. We review the anatomy, physiology, and cellular signaling of this system, and compare it to similar signaling in other organs/tissues of the body.


Subject(s)
Cochlea/pathology , Cochlea/physiology , Corticotropin-Releasing Hormone/metabolism , Hearing Loss, Noise-Induced/pathology , Hearing Loss, Noise-Induced/prevention & control , Signal Transduction/physiology , Animals , Auditory Threshold/physiology , Cochlea/cytology , Connexins/metabolism , Corticotropin-Releasing Hormone/genetics , Glutamate-Ammonia Ligase/metabolism , Hair Cells, Auditory/cytology , Hair Cells, Auditory/pathology , Hair Cells, Auditory/physiology , Humans , Hypothalamo-Hypophyseal System/physiology , Pituitary-Adrenal System/physiology , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Urocortins/metabolism
17.
J Neurosci ; 31(4): 1267-78, 2011 Jan 26.
Article in English | MEDLINE | ID: mdl-21273411

ABSTRACT

Cells of the inner ear face constant metabolic and structural stress. Exposure to intense sound or certain drugs destroys cochlea hair cells, which in mammals do not regenerate. Thus, an endogenous stress response system may exist within the cochlea to protect it from everyday stressors. We recently described the existence of corticotropin-releasing factor (CRF) in the mouse cochlea. The CRF receptor type 1 (CRFR1) is considered the primary and canonical target of CRF signaling, and systemically it plays an essential role in coordinating the body-wide stress response via activation of the hypothalamic-pituitary-adrenal (HPA) axis. Here, we describe an essential role for CRFR1 in auditory system development and function, and offer the first description of a complete HPA equivalent signaling system resident within the cochlea. To reveal the role of CRFR1 activation in the cochlea, we have used mice carrying a null ablation of the CRFR1 gene. CRFR1(-/-) mice exhibited elevated auditory thresholds at all frequencies tested, indicating reduced sensitivity. Furthermore, our results suggest that CRFR1 has a developmental role affecting inner hair cell morphology and afferent and efferent synapse distribution. Given the role of HPA signaling in maintaining local homeostasis in other tissues, the presence of a cochlear HPA signaling system suggests important roles for CRFR1 activity in setting cochlear sensitivity, perhaps both neural and non-neural mechanisms. These data highlight the complex pleiotropic mechanisms modulated by CRFR1 signaling in the cochlea.


Subject(s)
Cochlea/physiology , Hair Cells, Auditory/physiology , Hypothalamo-Hypophyseal System/physiology , Pituitary-Adrenal System/physiology , Receptors, Corticotropin-Releasing Hormone/physiology , Adrenocorticotropic Hormone/biosynthesis , Animals , Auditory Threshold , Cochlea/innervation , Corticosterone/pharmacology , Corticotropin-Releasing Hormone/biosynthesis , Mice , Mice, Knockout , Neural Pathways , Paracrine Communication , Pro-Opiomelanocortin/biosynthesis , Receptor, Melanocortin, Type 2/biosynthesis , Receptors, Corticotropin-Releasing Hormone/biosynthesis , Receptors, Corticotropin-Releasing Hormone/genetics , Signal Transduction
18.
Pac Symp Biocomput ; : 50-61, 2011.
Article in English | MEDLINE | ID: mdl-21121032

ABSTRACT

Gene set analyses have become a standard approach for increasing the sensitivity of transcriptomic studies. However, analytical methods incorporating gene sets require the availability of pre-defined gene sets relevant to the underlying physiology being studied. For novel physiological problems, relevant gene sets may be unavailable or existing gene set databases may bias the results towards only the best-studied of the relevant biological processes. We describe a successful attempt to mine novel functional gene sets for translational projects where the underlying physiology is not necessarily well characterized in existing annotation databases. We choose targeted training data from public expression data repositories and define new criteria for selecting biclusters to serve as candidate gene sets. Many of the discovered gene sets show little or no enrichment for informative Gene Ontology terms or other functional annotation. However, we observe that such gene sets show coherent differential expression in new clinical test data sets, even if derived from different species, tissues, and disease states. We demonstrate the efficacy of this method on a human metabolic data set, where we discover novel, uncharacterized gene sets that are diagnostic of diabetes, and on additional data sets related to neuronal processes and human development. Our results suggest that our approach may be an efficient way to generate a collection of gene sets relevant to the analysis of data for novel clinical applications where existing functional annotation is relatively incomplete.


Subject(s)
Data Mining/methods , Gene Regulatory Networks , Algorithms , Animals , Central Nervous System/metabolism , Cluster Analysis , Computational Biology , Databases, Genetic/statistics & numerical data , Female , Gene Expression Profiling/statistics & numerical data , Gene Expression Regulation, Developmental , Genome-Wide Association Study/statistics & numerical data , Humans , Male , Metabolic Diseases/genetics , Mice , Multigene Family , Oligonucleotide Array Sequence Analysis/statistics & numerical data , Pregnancy
19.
J Neurosci Res ; 88(13): 2976-90, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20544827

ABSTRACT

Generation of reactive oxygen species (ROS) is a common denominator in many conditions leading to cell death in the cochlea, yet little is known of the cochlea's endogenous mechanisms involved in preventing oxidative stress and its consequences in the cochlea. We have recently described a corticotropin-releasing factor (CRF) signaling system in the inner ear involved in susceptibility to noise-induced hearing loss. We use biochemical and proteomics assays to define further the role of CRF signaling in the response of cochlear cells to aminoglycoside exposure. We demonstrate that activity via the CRF(2) class of receptors protects against aminoglycoside-induced ROS production and activation of cell death pathways. This study suggests for the first time a role for CRF signaling in protecting the cochlea against oxidative stress, and our proteomics data suggest novel mechanisms beyond induction of free radical scavengers that are involved in its protective mechanisms.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Gentamicins/toxicity , Neurons/drug effects , Oxidative Stress/drug effects , Protein Synthesis Inhibitors/toxicity , Animals , Caspase 3/metabolism , Cell Death , Cell Line, Transformed , Ear, Inner/cytology , Gas Chromatography-Mass Spectrometry/methods , Proteome/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Superoxide Dismutase/metabolism
20.
J Neurosci ; 30(19): 6751-62, 2010 May 12.
Article in English | MEDLINE | ID: mdl-20463237

ABSTRACT

Acetylcholine is the major neurotransmitter of the olivocochlear efferent system, which provides feedback to cochlear hair cells and sensory neurons. To study the role of cochlear muscarinic receptors, we studied receptor localization with immunohistochemistry and reverse transcription-PCR and measured olivocochlear function, cochlear responses, and histopathology in mice with targeted deletion of each of the five receptor subtypes. M2, M4, and M5 were detected in microdissected immature (postnatal days 10-13) inner hair cells and spiral ganglion cells but not outer hair cells. In the adult (6 weeks), the same transcripts were found in microdissected organ of Corti and spiral ganglion samples. M2 protein was found, by immunohistochemistry, in olivocochlear fibers in both outer and inner hair cell areas. M3 mRNA was amplified only from whole cochleas, and M1 message was never seen in wild-type ears. Auditory brainstem responses (ABRs) and distortion product otoacoustic emissions (DPOAEs) were unaffected by loss of Gq-coupled receptors (M1, M3, or M5), as were shock-evoked olivocochlear effects and vulnerability to acoustic injury. In contrast, loss of Gi-coupled receptors (M2 and/or M4) decreased neural responses without affecting DPOAEs (at low frequencies). This phenotype and the expression pattern are consistent with excitatory muscarinic signaling in cochlear sensory neurons. At high frequencies, both ABRs and DPOAEs were attenuated by loss of M2 and/or M4, and the vulnerability to acoustic injury was dramatically decreased. This aspect of the phenotype and the expression pattern are consistent with a presynaptic role for muscarinic autoreceptors in decreasing ACh release from olivocochlear terminals during high-level acoustic stimulation and suggest that muscarinic antagonists could enhance the resistance of the inner ear to noise-induced hearing loss.


Subject(s)
Cochlea/physiology , Neurons, Afferent/physiology , Neurons, Efferent/physiology , Presynaptic Terminals/physiology , Receptors, Muscarinic/metabolism , Animals , Auditory Threshold/physiology , Cochlea/growth & development , Evoked Potentials, Auditory, Brain Stem , Feedback, Physiological , Female , Hair Cells, Auditory, Inner/physiology , Hair Cells, Auditory, Outer/physiology , Hearing Loss, Noise-Induced/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Otoacoustic Emissions, Spontaneous , RNA, Messenger/metabolism , Receptors, Muscarinic/genetics , Spiral Ganglion/growth & development , Spiral Ganglion/physiology , Synaptic Transmission/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...