Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
J Acoust Soc Am ; 153(1): 711, 2023 01.
Article in English | MEDLINE | ID: mdl-36732240

ABSTRACT

A series of articles discussing advanced diagnostics that can be used to assess noise injury and associated noise-induced hearing disorders (NIHD) was developed under the umbrella of the United States Department of Defense Hearing Center of Excellence Pharmaceutical Interventions for Hearing Loss working group. The overarching goals of the current series were to provide insight into (1) well-established and more recently developed metrics that are sensitive for detection of cochlear pathology or diagnosis of NIHD, and (2) the tools that are available for characterizing individual noise hazard as personal exposure will vary based on distance to the sound source and placement of hearing protection devices. In addition to discussing the utility of advanced diagnostics in patient care settings, the current articles discuss the selection of outcomes and end points that can be considered for use in clinical trials investigating hearing loss prevention and hearing rehabilitation.


Subject(s)
Deafness , Hearing Loss, Noise-Induced , Humans , Hearing Loss, Noise-Induced/diagnosis , Hearing Loss, Noise-Induced/etiology , Hearing Loss, Noise-Induced/prevention & control , Noise , Cochlea
2.
J Acoust Soc Am ; 151(4): 2802, 2022 04.
Article in English | MEDLINE | ID: mdl-35461487

ABSTRACT

Accumulating evidence suggests that cochlear deafferentation may contribute to suprathreshold deficits observed with or without elevated hearing thresholds, and can lead to accelerated age-related hearing loss. Currently there are no clinical diagnostic tools to detect human cochlear deafferentation in vivo. Preclinical studies using a combination of electrophysiological and post-mortem histological methods clearly demonstrate cochlear deafferentation including myelination loss, mitochondrial damages in spiral ganglion neurons (SGNs), and synaptic loss between inner hair cells and SGNs. Since clinical diagnosis of human cochlear deafferentation cannot include post-mortem histological quantification, various attempts based on functional measurements have been made to detect cochlear deafferentation. So far, those efforts have led to inconclusive results. Two major obstacles to the development of in vivo clinical diagnostics include a lack of standardized methods to validate new approaches and characterize the normative range of repeated measurements. In this overview, we examine strategies from previous studies to detect cochlear deafferentation from electrocochleography and auditory brainstem responses. We then summarize possible approaches to improve these non-invasive functional methods for detecting cochlear deafferentation with a focus on cochlear synaptopathy. We identify conceptual approaches that should be tested to associate unique electrophysiological features with cochlear deafferentation.


Subject(s)
Audiometry, Evoked Response , Evoked Potentials, Auditory, Brain Stem , Auditory Threshold/physiology , Cochlea , Evoked Potentials, Auditory, Brain Stem/physiology , Hearing , Humans , Spiral Ganglion
3.
J Acoust Soc Am ; 152(5): 2828, 2022 11.
Article in English | MEDLINE | ID: mdl-36456290

ABSTRACT

In 2019, the U.S. Food and Drug Administration issued guidance to increase the efficiency of drug development and support precision medicine, including tailoring treatments to those patients who will benefit based on genetic variation even in the absence of a documented mechanism of action. Although multiple advancements have been made in the field of pharmacogenetics (PGx) for other disease conditions, there are no approved PGx guidelines in the treatment of hearing disorders. In studies of noise-induced hearing loss (NIHL), some progress has been made in the last several years associating genomic loci with susceptibility to noise damage. However, the power of such studies is limited as the underlying physiological responses may vary considerably among the patient populations. Here, we have summarized previous animal studies to argue that NIHL subtyping is a promising strategy to increase the granularity of audiological assessments. By coupling this enhanced phenotyping capability with genetic association studies, we suggest that drug efficacy will be better predicted, increasing the likelihood of success in clinical trials when populations are stratified based on genetic variation or designed with multidrug combinations to reach a broader segment of individuals suffering or at risk from NIHL.


Subject(s)
Hearing Loss, Noise-Induced , Pharmacogenetics , United States , Animals , Precision Medicine , Hearing Loss, Noise-Induced/drug therapy , Hearing Loss, Noise-Induced/genetics , Genomics , Probability
4.
J Neurosci Res ; 98(9): 1800-1814, 2020 09.
Article in English | MEDLINE | ID: mdl-32415883

ABSTRACT

Deleterious age-related changes in the central auditory nervous system have been referred to as central age-related hearing impairment (ARHI) or central presbycusis. Central ARHI is often assumed to be the consequence of peripheral ARHI. However, it is possible that certain aspects of central ARHI are independent from peripheral ARHI. A confirmation of this possibility could lead to significant improvements in current rehabilitation practices. The major difficulty in addressing this issue arises from confounding factors, such as other age-related changes in both the cochlea and central non-auditory brain structures. Because gap detection is a common measure of central auditory temporal processing, and gap detection thresholds are less influenced by changes in other brain functions such as learning and memory, we investigated the potential relationship between age-related peripheral hearing loss (i.e., audiograms) and age-related changes in gap detection. Consistent with previous studies, a significant difference was found for gap detection thresholds between young and older adults. However, among older adults, no significant associations were observed between gap detection ability and several other independent variables including the pure tone audiogram average, the Wechsler Adult Intelligence Scale-Vocabulary score, gender, and age. Statistical analyses showed little or no contributions from these independent variables to gap detection thresholds. Thus, our data indicate that age-related decline in central temporal processing is largely independent of peripheral ARHI.


Subject(s)
Auditory Perception/physiology , Hearing Loss, Central/physiopathology , Presbycusis/physiopathology , Adult , Age Factors , Aged , Aged, 80 and over , Aging/physiology , Auditory Threshold , Cochlea/physiopathology , Female , Humans , Male , Middle Aged , Young Adult
5.
Pflugers Arch ; 466(4): 757-65, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24563219

ABSTRACT

T-type calcium channels are expressed in many diverse tissues, including neuronal, cardiovascular, and endocrine. T-type calcium channels are known to play roles in the development, maintenance, and repair of these tissues but have also been implicated in disease when not properly regulated. Calcium channel blockers have been developed to treat various diseases and their use clinically is widespread due to both their efficacy as well as their safety. Aside from their established clinical applications, recent studies have suggested neuroprotective effects of T-type calcium channel blockers. Many of the current T-type calcium channel blockers could act on other molecular targets besides T-type calcium channels making it uncertain whether their neuroprotective effects are solely due to blocking of T-type calcium channels. In this review, we discuss these drugs as well as newly developed chemical compounds that are designed to be more selective for T-type calcium channels. We review in vitro and in vivo evidence of neuroprotective effects by these T-type calcium channel blockers. We conclude by discussing possible molecular mechanisms underlying the neuroprotective effects by T-type calcium channel blockers.


Subject(s)
Calcium Channel Blockers/metabolism , Calcium Channels, T-Type/metabolism , Nervous System Diseases/metabolism , Nervous System Diseases/prevention & control , Neuroprotective Agents/metabolism , Animals , Calcium Channel Blockers/therapeutic use , Humans , Neuroprotective Agents/therapeutic use
6.
Stem Cells ; 30(8): 1756-70, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22714950

ABSTRACT

Mesenchymal stem cells (MSCs) exhibit extensive self-renewal potential and can modulate immunocyte activation. Our previous study reported that miR-181a expression was significantly increased in placenta from women with severe preeclampsia (PE), but the mechanisms by which miR-181a regulates MSCs are unknown. In this study, we asked if and how miR-181a regulates MSCs' proliferation and immunosuppressive properties. We found that the expression of miR-181a in the MSCs derived from the umbilical cord and decidua of PE patients increased relative to MSCs derived from normal patients. Transfection with miR-181a oligos prevented MSCs proliferation but did not affect MSCs apoptosis. Overexpression of miR-181a blocked activation of the TGF-ß signaling pathway and caused downregulation of target gene (TGFBR1 and TGFBRAP1) mRNA and protein expression. Reporter genes with putative miR-181a binding sites from the TGFBR1 and TGFBRAP1 3'-untranslated regions (3'-UTRs) were downregulated in the presence of miR-181a, suggesting that miR-181a binds to TGFBR1 and TGFBRAP1 3'-UTRs. In contrast, transfection of MSCs with miR-181a oligo enhanced expression of IL-6 and indoleamine 2,3-dioxygenase by activating p38 and JNK signaling pathways, respectively. MSCs transfected with miR-181a also enhanced the proliferation of T cells in a short-term culture. Additionally, treatment with control MSCs, but not miR-181a transfected MSCs, improved dextran sulfate sodium-induced experimental colitis, suggesting that miR-181a attenuates the immunosuppressive properties of MSCs in vivo. Together, our data demonstrate that miR-181a is an important endogenous regulator in the proliferation and immunosuppressive properties of MSCs.


Subject(s)
Mesenchymal Stem Cells/immunology , MicroRNAs/immunology , Animals , Cell Growth Processes/immunology , Disease Models, Animal , Humans , Interleukin-6/immunology , Mesenchymal Stem Cells/cytology , Mice , MicroRNAs/genetics , Transfection , Transforming Growth Factor beta/immunology
7.
Stem Cells ; 29(12): 1963-74, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21956995

ABSTRACT

Induced pluripotent stem cells (iPSCs) can be reprogrammed from adult somatic cells by transduction with Oct4, Sox2, Klf4, and c-Myc, but the molecular cascades initiated by these factors remain poorly understood. Impeding their elucidation is the stochastic nature of the iPS induction process, which results in heterogeneous cell populations. Here we have synchronized the reprogramming process by a two-phase induction: an initial stable intermediate phase following transduction with Oct4, Klf4, and c-Myc, and a final iPS phase following overexpression of Sox2. This approach has enabled us to examine temporal gene expression profiles, permitting the identification of Sox2 downstream genes critical for induction. Furthermore, we have validated the feasibility of our new approach by using it to confirm that downregulation of transforming growth factor ß signaling by Sox2 proves essential to the reprogramming process. Thus, we present a novel means for dissecting the details underlying the induction of iPSCs, an approach with significant utility in this arena and the potential for wide-ranging implications in the study of other reprogramming mechanisms.


Subject(s)
Induced Pluripotent Stem Cells/cytology , SOXB1 Transcription Factors/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Cell Differentiation , Cells, Cultured/cytology , Female , Fibroblasts/cytology , Gene Expression Profiling , Gene Expression Regulation , Immunohistochemistry , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Mice, Nude , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Plasmids/genetics , Plasmids/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Retroviridae/genetics , Retroviridae/metabolism , SOXB1 Transcription Factors/genetics , Teratoma/genetics , Teratoma/metabolism , Teratoma/pathology , Transforming Growth Factor beta/genetics
8.
Gerontology ; 58(6): 490-6, 2012.
Article in English | MEDLINE | ID: mdl-22710288

ABSTRACT

Hearing loss is a common age-associated affliction that can result from the loss of hair cells and spiral ganglion neurons (SGNs) in the cochlea. Although hair cells and SGNs are typically lost in the same cochlea, recent analysis suggests that they can occur independently, via unique mechanisms. Research has identified both environmental and genetic factors that contribute to degeneration of cochlear cells. Additionally, molecular analysis has identified multiple cell-signaling mechanisms that likely contribute to pathological changes that result in hearing deficiencies. These analyses should serve as useful primers for future work, including genomic and proteomic analysis, to elucidate the mechanisms driving cell loss in the aging cochlea. Significant progress in this field has occurred in the past decade. As our understanding of aging-induced cochlear changes continues to improve, our ability to offer medical intervention will surely benefit the growing elderly population.


Subject(s)
Presbycusis , Aged , Animals , Calcium Signaling , Cell Death , Disease Models, Animal , Female , Glucocorticoids/metabolism , Glutamic Acid/metabolism , Gonadal Steroid Hormones/metabolism , Hair Cells, Auditory/pathology , Hair Cells, Auditory/physiology , Hearing Loss, Noise-Induced/etiology , Humans , Male , Middle Aged , Noise/adverse effects , Oxidative Stress , Presbycusis/etiology , Presbycusis/pathology , Presbycusis/physiopathology , Signal Transduction , Spiral Ganglion/pathology , Spiral Ganglion/physiopathology , Stress, Physiological
9.
Front Cell Neurosci ; 16: 851500, 2022.
Article in English | MEDLINE | ID: mdl-35356798

ABSTRACT

The sound-evoked electrical compound potential known as auditory brainstem response (ABR) represents the firing of a heterogenous population of auditory neurons in response to sound stimuli, and is often used for clinical diagnosis based on wave amplitude and latency. However, recent ABR applications to detect human cochlear synaptopathy have led to inconsistent results, mainly due to the high variability of ABR wave-1 amplitude. Here, rather than focusing on the amplitude of ABR wave 1, we evaluated the use of ABR wave curvature to detect cochlear synaptic loss. We first compared four curvature quantification methods using simulated ABR waves, and identified that the cubic spline method using five data points produced the most accurate quantification. We next evaluated this quantification method with ABR data from an established mouse model with cochlear synaptopathy. The data clearly demonstrated that curvature measurement is more sensitive and consistent in identifying cochlear synaptic loss in mice compared to the amplitude and latency measurements. We further tested this curvature method in a different mouse model presenting with otitis media. The change in curvature profile due to middle ear infection in otitis media is different from the profile of mice with cochlear synaptopathy. Thus, our study suggests that curvature quantification can be used to address the current ABR variability issue, and may lead to additional applications in the clinic diagnosis of hearing disorders.

10.
J Neurosci ; 30(50): 17068-78, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21159977

ABSTRACT

The vast majority of Alzheimer's disease (AD) cases are late onset with progressive synapse loss and neurodegeneration. Although the amyloid hypothesis has generated great insights into the disease mechanism, several lines of evidence indicate that other risk factors might precondition the brain to amyloid toxicity. Here, we show that the deletion of a major lipoprotein receptor, low-density lipoprotein receptor-related protein 1 (LRP1), in forebrain neurons in mice leads to a global defect in brain lipid metabolism characterized by decreased brain levels of cholesterol, sulfatide, galactosylceramide, and triglyceride. These lipid deficits correlate with progressive, age-dependent dendritic spine degeneration, synapse loss, neuroinflammation, memory loss, and eventual neurodegeneration. We further show that the levels of glutamate receptor subunits NMDA receptor 1 and Glu receptor 1 are selectively reduced in LRP1 forebrain knock-out mice and in LRP1 knockdown neurons, which is partially rescued by restoring neuronal cholesterol. Together, these studies support a critical role for LRP1 in maintaining brain lipid homeostasis and associated synaptic and neuronal integrity, and provide important insights into the pathophysiological mechanisms in AD.


Subject(s)
Lipid Metabolism/genetics , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Nerve Degeneration/metabolism , Neurons/pathology , Prosencephalon/metabolism , Synapses/pathology , Age Factors , Amnesia/pathology , Animals , Cell Culture Techniques , Dendritic Spines/pathology , Hippocampus/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Mice , Mice, Knockout , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Neurons/metabolism , Prosencephalon/pathology , Receptors, AMPA/biosynthesis , Receptors, N-Methyl-D-Aspartate/biosynthesis , Synapses/metabolism
11.
J Biol Chem ; 285(20): 14909-14919, 2010 May 14.
Article in English | MEDLINE | ID: mdl-20228067

ABSTRACT

Mutations in the COCH (coagulation factor C homology) gene have been attributed to DFNA9 (deafness, autosomal-dominant 9), an autosomal-dominant non-syndromic hearing loss disorder. However, the mechanisms responsible for DFNA9 hearing loss remain unknown. Here, we demonstrate that mutant cochlin, the protein product of the COCH gene, forms a stable dimer that is sensitive to reducing agent. In contrast, wild-type (WT) cochlin may form only dimers transiently. Interestingly, the presence of mutant cochlin can stabilize WT cochlin in dimer conformation, providing a possible mechanism for the dominant nature of DFNA9 mutations. Furthermore, the expression of mutant cochlin eventually induces WT cochlin to form stable oligomers that are resistant to reducing agent. Finally, we show that mutant cochlin is cytotoxic in vitro and in vivo. Our study suggests a possible molecular mechanism underlying DFNA9 hearing loss and provides an in vitro model that may be used to explore protein-misfolding diseases in general.


Subject(s)
Hearing Loss/genetics , Protein Folding , Proteins/genetics , Animals , Blotting, Western , Cell Line , Electrophoresis, Polyacrylamide Gel , Extracellular Matrix Proteins , Humans , Immunoprecipitation , Mice , Microscopy, Fluorescence , Point Mutation
12.
J Assoc Res Otolaryngol ; 22(2): 107-126, 2021 04.
Article in English | MEDLINE | ID: mdl-33415542

ABSTRACT

Cisplatin chemotherapy often causes permanent hearing loss, which leads to a multifaceted decrease in quality of life. Identification of early cisplatin-induced cochlear damage would greatly improve clinical diagnosis and provide potential drug targets to prevent cisplatin's ototoxicity. With improved functional and immunocytochemical assays, a recent seminal discovery revealed that synaptic loss between inner hair cells and spiral ganglion neurons is a major form of early cochlear damage induced by noise exposure or aging. This breakthrough discovery prompted the current study to determine early functional, cellular, and molecular changes for cisplatin-induced hearing loss, in part to determine if synapse injury is caused by cisplatin exposure. Cisplatin was delivered in one to three treatment cycles to both male and female mice. After the cisplatin treatment of three cycles, threshold shift was observed across frequencies tested like previous studies. After the treatment of two cycles, beside loss of outer hair cells and an increase in high-frequency hearing thresholds, a significant latency delay of auditory brainstem response wave 1 was observed, including at a frequency region where there were no changes in hearing thresholds. The wave 1 latency delay was detected as early cisplatin-induced ototoxicity after only one cycle of treatment, in which no significant threshold shift was found. In the same mice, mitochondrial loss in the base of the cochlea and declining mitochondrial morphometric health were observed. Thus, we have identified early spiral ganglion-associated functional and cellular changes after cisplatin treatment that precede significant threshold shift.


Subject(s)
Cisplatin , Cochlea , Deafness , Ototoxicity , Animals , Cisplatin/toxicity , Cochlea/drug effects , Cochlea/physiopathology , Deafness/chemically induced , Female , Hearing , Male , Mice
13.
J Neurosci ; 29(7): 2022-6, 2009 Feb 18.
Article in English | MEDLINE | ID: mdl-19228956

ABSTRACT

Methylprednisolone (MP), a synthetic glucocorticoid agonist, is widely used for the clinical therapy of white matter diseases in the nervous system, such as spinal cord injury and multiple sclerosis. In addition to its potent anti-inflammatory and antioxidant properties, we recently discovered a selective antiapoptotic effect of MP on oligodendrocytes via the activation of the glucocorticoid receptor (GR) and the upregulation of bcl-X(L), a splicing isoform of the bcl-x gene. Based on published findings of the functional interactions between GR and STAT5, a transcription factor from the family of signal transducers and activators of transcription (STAT), we examined whether the glucocorticoid signaling pathway interacts with STAT5 to upregulate bcl-X(L) and protect oligodendrocytes. We show herein that (1) the GR and STAT5 complex is present on the STAT5-binding site of the bcl-x promoter region in oligodendrocytes; (2) the overexpression of an activated form of STAT5 prevents alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-induced oligodendrocyte cell death; and (3) this prevention is lost when the STAT5 gene is knocked down. Thus, our results provide one molecular mechanism underlying the postinjury protective effects of oligodendrocytes by stress hormones.


Subject(s)
Apoptosis/drug effects , Methylprednisolone/pharmacology , Oligodendroglia/drug effects , Oligodendroglia/metabolism , STAT5 Transcription Factor/drug effects , STAT5 Transcription Factor/metabolism , Animals , Animals, Newborn , Apoptosis/genetics , Binding Sites/drug effects , Binding Sites/genetics , Cells, Cultured , Cytoprotection/drug effects , Cytoprotection/genetics , Down-Regulation/genetics , Multiple Sclerosis/drug therapy , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Neuroprotective Agents/pharmacology , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , RNA Interference , Rats , Receptors, Glucocorticoid/drug effects , Receptors, Glucocorticoid/metabolism , STAT5 Transcription Factor/genetics , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/genetics , Spinal Cord Injuries/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology , bcl-X Protein/drug effects , bcl-X Protein/metabolism
14.
J Cell Biol ; 161(6): 1133-41, 2003 Jun 23.
Article in English | MEDLINE | ID: mdl-12821646

ABSTRACT

Transmembrane isoforms of neuregulin-1 (Nrg-1), ligands for erbB receptors, include an extracellular domain with an EGF-like sequence and a highly conserved intracellular domain (ICD) of unknown function. In this paper, we demonstrate that transmembrane isoforms of Nrg-1 are bidirectional signaling molecules in neurons. The stimuli for Nrg-1 back signaling include binding of erbB receptor dimers to the extracellular domain of Nrg-1 and neuronal depolarization. These stimuli elicit proteolytic release and translocation of the ICD of Nrg-1 to the nucleus. Once in the nucleus, the Nrg-1 ICD represses expression of several regulators of apoptosis, resulting in decreased neuronal cell death in vitro. Thus, regulated proteolytic processing of Nrg-1 results in retrograde signaling that appears to mediate contact and activity-dependent survival of Nrg-1-expressing neurons.


Subject(s)
Cell Membrane/metabolism , Cell Survival/physiology , Neuregulin-1/metabolism , Neurons, Afferent/metabolism , Signal Transduction/physiology , Animals , Apoptosis/physiology , Cell Communication/physiology , Cell Membrane/ultrastructure , Cells, Cultured , ErbB Receptors/metabolism , Membrane Potentials/physiology , Mice , Microscopy, Electron , Neurons, Afferent/ultrastructure , Peptide Hydrolases/metabolism , Protein Structure, Tertiary/physiology , Protein Transport/physiology , Spiral Ganglion/metabolism , Spiral Ganglion/ultrastructure
15.
Pharmacol Ther ; 198: 206-219, 2019 06.
Article in English | MEDLINE | ID: mdl-30831129

ABSTRACT

Granulocytes are the major type of phagocytes constituting the front line of innate immune defense against bacterial infection. In adults, granulocytes are derived from hematopoietic stem cells in the bone marrow. Alcohol is the most frequently abused substance in human society. Excessive alcohol consumption injures hematopoietic tissue, impairing bone marrow production of granulocytes through disrupting homeostasis of granulopoiesis and the granulopoietic response. Because of the compromised immune defense function, alcohol abusers are susceptible to infectious diseases, particularly septic infection. Alcoholic patients with septic infection and granulocytopenia have an exceedingly high mortality rate. Treatment of serious infection in alcoholic patients with bone marrow inhibition continues to be a major challenge. Excessive alcohol consumption also causes diseases in other organ systems, particularly severe alcoholic hepatitis which is life threatening. Corticosteroids are the only therapeutic option for improving short-term survival in patients with severe alcoholic hepatitis. The existence of advanced alcoholic liver diseases and administration of corticosteroids make it more difficult to treat serious infection in alcoholic patients with the disorder of granulopoieis. This article reviews the recent development in understanding alcohol-induced disruption of marrow granulopoiesis and the granulopoietic response with the focus on progress in delineating cell signaling mechanisms underlying the alcohol-induced injury to hematopoietic tissue. Efforts in exploring effective therapy to improve patient care in this field will also be discussed.


Subject(s)
Agranulocytosis/etiology , Alcoholism/complications , Ethanol/adverse effects , Granulocytes/drug effects , Hematopoietic Stem Cells/drug effects , Leukopoiesis/drug effects , Animals , Homeostasis/drug effects , Humans
16.
Nat Neurosci ; 7(11): 1250-8, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15494726

ABSTRACT

Neuregulin-1 (Nrg-1) contains an intracellular domain (Nrg-ICD) that translocates into the nucleus, where it may regulate gene expression upon neuronal depolarization. However, the identity of its target promoters and the mechanisms by which it regulates transcription have been elusive. Here we report that, in the mouse cochlea, synaptic activity increases the level of nuclear Nrg-ICD and upregulates postsynaptic density protein-95 (PSD-95), a scaffolding protein that is enriched in post-synaptic structures. Nrg-ICD enhances the transcriptional activity of the PSD-95 promoter by binding to a zinc-finger transcription factor, Eos. The Nrg-ICD-Eos complex induces endogenous PSD-95 expression in vivo through a signaling pathway that is mostly independent of gamma-secretase regulation. This upregulation of PSD-95 expression by the Nrg-ICD-Eos complex provides a molecular basis for activity-dependent synaptic plasticity.


Subject(s)
Carrier Proteins/physiology , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Neuregulin-1/physiology , Neurons/physiology , Transcription, Genetic/physiology , Acoustic Stimulation/methods , Animals , Cell Line , Cochlea/cytology , Cochlea/physiology , Cochlea/radiation effects , DNA-Binding Proteins/physiology , Disks Large Homolog 4 Protein , Electrophoretic Mobility Shift Assay/methods , Extracellular Space/metabolism , Extracellular Space/radiation effects , Gene Expression Regulation/radiation effects , Genes, Reporter/physiology , Guanylate Kinases , Humans , Ikaros Transcription Factor , Immunohistochemistry/methods , Immunoprecipitation/methods , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Mice , Mutagenesis , Nerve Tissue Proteins/genetics , Neuronal Plasticity/physiology , Neuronal Plasticity/radiation effects , Neurons/cytology , Neurons/radiation effects , Oligonucleotide Array Sequence Analysis/methods , Potassium Chloride/pharmacology , Promoter Regions, Genetic/physiology , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction/methods , Signal Transduction/physiology , Subcellular Fractions/metabolism , Time Factors , Transcription Factors/physiology , Transfection/methods
17.
Hear Res ; 367: 88-96, 2018 09.
Article in English | MEDLINE | ID: mdl-30071403

ABSTRACT

Gene expression analysis is essential for understanding the rich repertoire of cellular functions. With the development of sensitive molecular tools such as single-cell RNA sequencing, extensive gene expression data can be obtained and analyzed from various tissues. Single-molecule fluorescence in situ hybridization (smFISH) has emerged as a powerful complementary tool for single-cell genomics studies because of its ability to map and quantify the spatial distributions of single mRNAs at the subcellular level in their native tissue. Here, we present a detailed method to study the copy numbers and spatial localizations of single mRNAs in the cochlea and inferior colliculus. First, we demonstrate that smFISH can be performed successfully in adult cochlear tissue after decalcification. Second, we show that the smFISH signals can be detected with high specificity. Third, we adapt an automated transcript analysis pipeline to quantify and identify single mRNAs in a cell-specific manner. Lastly, we show that our method can be used to study possible correlations between transcriptional and translational activities of single genes. Thus, we have developed a detailed smFISH protocol that can be used to study the expression of single mRNAs in specific cell types of the peripheral and central auditory systems.


Subject(s)
Auditory Pathways/metabolism , Cochlea/metabolism , In Situ Hybridization, Fluorescence , Inferior Colliculi/metabolism , Neurons/metabolism , RNA, Messenger/genetics , Single-Cell Analysis/methods , Animals , Auditory Pathways/cytology , Cochlea/cytology , Gene Expression Regulation , Immunohistochemistry , Inferior Colliculi/cytology , Mice , Microscopy, Confocal , Neurons/cytology , RNA, Messenger/metabolism , Transcription, Genetic
18.
J Assoc Res Otolaryngol ; 19(6): 653-668, 2018 12.
Article in English | MEDLINE | ID: mdl-30187298

ABSTRACT

Noise is the most common occupational and environmental hazard, and noise-induced hearing loss (NIHL) is the second most common form of sensorineural hearing deficit. Although therapeutics that target the free-radical pathway have shown promise, none of these compounds is currently approved against NIHL by the United States Food and Drug Administration. The present study has demonstrated that tetrandrine (TET), a traditional Chinese medicinal alkaloid and the main chemical isolate of the Stephania tetrandra S. Moore herb, significantly attenuated NIHL in CBA/CaJ mice. TET is known to exert antihypertensive and antiarrhythmic effects through the blocking of calcium channels. Whole-cell patch-clamp recording from adult spiral ganglion neurons showed that TET blocked the transient Ca2+ current in a dose-dependent manner and the half-blocking concentration was 0.6 + 0.1 µM. Consistent with previous findings that modulations of calcium-based signaling pathways have both prophylactic and therapeutic effects against neural trauma, NIHL was significantly diminished by TET administration. Importantly, TET has a long-lasting protective effect after noise exposure (48 weeks) in comparison to 2 weeks after noise exposure. The otoprotective effects of TET were achieved mainly by preventing outer hair cell damage and synapse loss between inner hair cells and spiral ganglion neurons. Thus, our data indicate that TET has great potential in the prevention and treatment of NIHL.


Subject(s)
Benzylisoquinolines/therapeutic use , Calcium Channel Blockers/therapeutic use , Hearing Loss, Noise-Induced/prevention & control , Phytotherapy , Stephania tetrandra , Animals , Benzylisoquinolines/analysis , Benzylisoquinolines/pharmacology , Calcium Channel Blockers/pharmacology , Drug Evaluation, Preclinical , Evoked Potentials, Auditory, Brain Stem/drug effects , Female , Male , Mice , Spiral Ganglion/drug effects
19.
Hear Res ; 226(1-2): 79-91, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17107766

ABSTRACT

The protective benefits of hypoxic preconditioning (HPC) against permanent noise-induced hearing loss (NIHL) were investigated in mice. Hypoxia induced by exposure to 8% O2 for 4 h conferred significant protection against damaging broadband noise delivered 24-48 h later in male and female CBA/J (CBA) and CBA/CaJ mice. No protection was found in C57BL/6 (B6) mice, their B6.CAST-Cdh23(CAST) (B6.CAST) congenics, or in CBAxB6 F1 hybrid mice over the same interval, suggesting that the potential for HPC depends on one or a few autosomal recessive alleles carried by CBA-related strains, and is not influenced by the Cdh23 locus. Protection against NIHL in CBA mice was associated with significant up-regulation of hypoxia-inducible factor-1alpha (HIF-1alpha) within the organ of Corti, not found in B6.CAST. In both CBA and B6.CAST mice, some hypoxia-noise intervals shorter than 24 h were associated with exacerbation of NIHL. Cellular cascades underlying the early exacerbation of NIHL by hypoxia are therefore common to both strains, and not mechanistically linked to later protection. Elucidation of the events that underlie HPC, and how these are impacted by genetics, may lead to pharmacologic approaches to mimic HPC, and may help identify individuals with elevated risk of NIHL.


Subject(s)
Hearing Loss, Noise-Induced/genetics , Hearing Loss, Noise-Induced/prevention & control , Ischemic Preconditioning/methods , Animals , Animals, Congenic , Cadherins/genetics , Cochlea/pathology , Evoked Potentials, Auditory, Brain Stem , Female , Hearing Loss, Noise-Induced/pathology , Hearing Loss, Noise-Induced/physiopathology , Hypoxia/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Organ of Corti/blood supply , Organ of Corti/physiopathology , Species Specificity , Time Factors , Up-Regulation
20.
Hear Res ; 226(1-2): 52-60, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17291698

ABSTRACT

Cochlear noise injury is the second most frequent cause of sensorineural hearing loss, after aging. Because calcium dysregulation is a widely recognized contributor to noise injury, we examined the potential of calcium channel blockers to reduce noise-induced hearing loss (NIHL) in mice. We focused on two T-type calcium blockers, trimethadione and ethosuximide, which are anti-epileptics approved by the Food and Drug Administration. Young C57BL/6 mice of either gender were divided into three groups: a 'prevention' group receiving the blocker via drinking water before noise exposure; a 'treatment' group receiving the blocker via drinking water after noise exposure; and controls receiving noise alone. Trimethadione significantly reduced NIHL when applied before noise exposure, as determined by auditory brainstem recording. Both ethosuximide and trimethadione were effective in reducing NIHL when applied after noise exposure. Results were influenced by gender, with males generally receiving greater benefit than females. Quantitation of hair cell and neuronal density suggested that preservation of outer hair cells could account for the observed protection. Immunocytochemistry and RT-PCR suggested that this protection involves direct action of T-type blockers on alpha1 subunits comprising one or more Ca(v)3 calcium channel types in the cochlea. Our findings provide a basis for clinical studies testing T-type calcium blockers both to prevent and treat NIHL.


Subject(s)
Calcium Channel Blockers/pharmacology , Ethosuximide/pharmacology , Hearing Loss, Noise-Induced/drug therapy , Hearing Loss, Noise-Induced/prevention & control , Trimethadione/pharmacology , Animals , Auditory Threshold/drug effects , Base Sequence , Calcium Channel Blockers/therapeutic use , Calcium Channels, T-Type/drug effects , Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/metabolism , Cochlea/drug effects , Cochlea/metabolism , Cochlea/pathology , DNA Primers/genetics , Ethosuximide/therapeutic use , Female , Hearing Loss, Noise-Induced/genetics , Hearing Loss, Noise-Induced/pathology , Male , Mice , Mice, Inbred C57BL , Sex Characteristics , Trimethadione/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL