1
|
Sato R, Adachi R, Yokoi N, Tsujimura K, Egawa R, Hara Y, Fukata Y, Fukata M, Ogi T, Sone M, Kuba H. Loss of neuronal activity facilitates surface accumulation of p75NTR and cell death in avian cochlear nucleus. Neurosci Res 2025; 213:23-34. [PMID: 39848466 DOI: 10.1016/j.neures.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 01/25/2025]
Abstract
Sensorineural hearing loss causes cell death in central auditory neurons, but molecular mechanisms of triggering this process are not fully understood. We report here that loss of afferent activity promotes cell death by facilitating proBDNF-p75NTR signals in cochlear nucleus of chicks around hatch. RNA-seq analyses revealed up-regulation of genes related to proBDNF-p75NTR-JNK signals as well as apoptosis at the nucleus within 24 h after unilateral cochlea deprivation. Western blotting confirmed a high level of proBDNF protein at the nucleus. Moreover, FLAG-tagged p75NTR accumulated at the plasma membrane of the neurons within 6 h after the deprivation, well before the upregulation of apoptotic genes. Cell viability assay using propidium iodide in organ culture showed that proBDNF increased the fraction of dying neurons in a dose-dependent manner. In addition, pharmacological blockades of synaptic and spike activities in the culture reproduced the surface accumulation of p75NTR in vivo and increased the fraction of dying neurons, while genetic inhibition of p75NTR signals occluded the cell death during the activity blockades. These results indicate that afferent activity is crucial for suppressing surface accumulation of p75NTR and hence proBDNF-p75NTR signals and that the loss of this suppression would contribute to triggering cell death after deafferentation in the developing brainstem auditory circuit.
Collapse
Affiliation(s)
- Ryosuke Sato
- Department of Cell Physiology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan; Department of Otorhinolaryngology Head and Neck Surgery, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Ryota Adachi
- Department of Cell Physiology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Norihiko Yokoi
- Division of Neuropharmacology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Keita Tsujimura
- Group of Brain Function and Development, Nagoya University Neuroscience Institute of the Graduate School of Science, Nagoya 464-8602, Japan; Research Unit for Developmental Disorders, Institute for Advanced Research, Nagoya University, Nagoya 464-8602, Japan
| | - Ryo Egawa
- Department of Cell Physiology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Yuichiro Hara
- Department of Genetics, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan; Department of Human Genetics and Molecular Biology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Yuko Fukata
- Division of Molecular and Cellular Pharmacology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Masaki Fukata
- Division of Neuropharmacology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Tomoo Ogi
- Department of Genetics, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan; Department of Human Genetics and Molecular Biology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Michihiko Sone
- Department of Otorhinolaryngology Head and Neck Surgery, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Hiroshi Kuba
- Department of Cell Physiology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan.
| |
Collapse
|
2
|
McLellan K, Sabbagh S, Takahashi M, Hong H, Wang Y, Sanchez JT. BDNF Differentially Affects Low- and High-Frequency Neurons in a Primary Nucleus of the Chicken Auditory Brainstem. BIOLOGY 2024; 13:877. [PMID: 39596832 PMCID: PMC11592191 DOI: 10.3390/biology13110877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/23/2024] [Accepted: 10/27/2024] [Indexed: 11/29/2024]
Abstract
Neurotrophins are proteins that mediate neuronal development using spatiotemporal signaling gradients. The chicken nucleus magnocellularis (NM), an analogous structure to the mammalian anteroventral cochlear nucleus, provides a model system in which signaling between the brain-derived neurotrophic factor (BDNF) and tyrosine receptor kinase B (TrkB) is temporally regulated. In the NM, TrkB expression is high early in development (embryonic [E] day 9) and is downregulated until maturity (E18-21). It is currently unknown how BDNF-TrkB signaling affects neuronal properties throughout development and across a spatial (i.e., frequency) axis. To investigate this, we exogenously applied BDNF onto NM neurons ex vivo and studied intrinsic properties using whole-cell patch clamp electrophysiology. Early in development (E13), when TrkB expression is detectable with immunohistochemistry, BDNF application slowed the firing of high-frequency NM neurons, resembling an immature phenotype. Current measurements and biophysical modeling revealed that this was mediated by a decreased conductance of the voltage-dependent potassium channels. Interestingly, this effect was seen only in high-frequency neurons and not in low-frequency neurons. BDNF-TrkB signaling induced minimal changes in late-developing NM neurons (E20-21) of high and low frequencies. Our results indicate that normal developmental downregulation of BDNF-TrkB signaling promotes neuronal maturation tonotopically in the auditory brainstem, encouraging the appropriate development of neuronal properties.
Collapse
Affiliation(s)
- Kristine McLellan
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL 60208, USA
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Sima Sabbagh
- Program in Neuroscience, Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
| | - Momoko Takahashi
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Hui Hong
- Oregon Hearing Research Center and Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Yuan Wang
- Program in Neuroscience, Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
| | - Jason Tait Sanchez
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL 60208, USA
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
- Knowles Hearing Research Center, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
3
|
Nagashima D, Mizukami N, Ogawa N, Suzuki S, Ohno M, Aoki R, Furukawa M, Izumo N. Bovine Lactoferrin Promotes Neurite Outgrowth in PC12 Cells via the TrkA Receptor. Int J Mol Sci 2024; 25:11249. [PMID: 39457031 PMCID: PMC11508191 DOI: 10.3390/ijms252011249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/12/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Lactoferrin (LF) is a multifunctional protein abundant in breast milk that modulates the functions of neural stem cells. Recent studies have demonstrated the efficacy of bovine LF (bLF) in mitigating behavioral changes; however, the molecular mechanisms on the nervous system have not yet been elucidated. The presented study aimed to characterize the molecular mechanisms of bLF on nerve extension in PC12 cells. PC12 cells were treated with 0.01-1000 µg/mL of bLF, and cell viability was determined using the cell counting kit-8 assay after treatment for 24 h. Morphometric evaluation was performed after 24 or 72 h of treatment with 50 ng/mL nerve growth factor (NGF) or 100-500 µg/mL bLF. The molecular mechanisms were investigated using Western blotting and real-time quantitative PCR. Cell viability was significantly decreased after treatment with 600-1000 µg/mL bLF for 24 h compared with the control group. Morphometric evaluation revealed neurite outgrowth after 72 h of NGF treatment, with a significant increase in neurite outgrowth after treatment with 250 µg/mL bLF. The phosphorylated p44/42 expression ratio peaked at 5 min and persisted for up to 10 min. Quantitative real-time PCR revealed a significant decrease in MAP2 expression. Our findings suggested that bLF enhanced PC12 cell neurite outgrowth to a similar extent as NGF. These effects are thought to be mediated via the TrkA receptor and activated by the phosphorylated ERK signaling pathway. Therefore, this study demonstrates that bLF promotes neurite outgrowth via a pathway similar to that of NGF.
Collapse
Affiliation(s)
- Daichi Nagashima
- Laboratory of Clinical Pharmaceutics, Yokohama University of Pharmacy, Yokohama 245-0066, Kanagawa, Japan;
- General Health Medical Research Center, Yokohama University of Pharmacy, Yokohama 245-0066, Kanagawa, Japan
| | - Noa Mizukami
- Laboratory of Pharmacotherapy, Yokohama University of Pharmacy, Yokohama 245-0066, Kanagawa, Japan
| | - Nana Ogawa
- Laboratory of Pharmacotherapy, Yokohama University of Pharmacy, Yokohama 245-0066, Kanagawa, Japan
| | - Sayaka Suzuki
- Laboratory of Pharmacotherapy, Yokohama University of Pharmacy, Yokohama 245-0066, Kanagawa, Japan
| | - Megumi Ohno
- NRL Pharma Inc., Kawasaki 213-0012, Kanagawa, Japan
| | - Ryoken Aoki
- Center for Pharmaceutical Education, Yokohama University of Pharmacy, Yokohama 245-0066, Kanagawa, Japan
| | - Megumi Furukawa
- Center for Pharmaceutical Education, Yokohama University of Pharmacy, Yokohama 245-0066, Kanagawa, Japan
| | - Nobuo Izumo
- General Health Medical Research Center, Yokohama University of Pharmacy, Yokohama 245-0066, Kanagawa, Japan
- Laboratory of Pharmacotherapy, Yokohama University of Pharmacy, Yokohama 245-0066, Kanagawa, Japan
| |
Collapse
|
4
|
Ordiway G, McDonnell M, Sanchez JT. Revisiting the Chicken Auditory Brainstem Response: Frequency Specificity, Threshold Sensitivity, and Cross Species Comparison. Neurosci Insights 2024; 19:26331055241228308. [PMID: 38304551 PMCID: PMC10832403 DOI: 10.1177/26331055241228308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 01/09/2024] [Indexed: 02/03/2024] Open
Abstract
The auditory brainstem response (ABR) is important for both clinical and basic auditory research. It is a non-invasive measure of hearing function with millisecond-level precision. The ABR can not only measure the synchrony, speed, and efficacy of auditory physiology but also detect different modalities of hearing pathology and hearing loss. ABRs are easily acquired in vertebrate animal models like reptiles, birds, and mammals, and complement existing molecular, developmental, and systems-level research. One such model system is the chicken; an excellent animal for studying auditory development, structure, and function. However, the ABR for chickens was last reported nearly 4 decades ago. The current study examines how decades of ABR characterization in other animal species support findings from the chicken ABR. We replicated and expanded on previous research using 43 chicken hatchlings 1- and 2-day post-hatch. We report that click-evoked chicken ABRs presented with a peak waveform morphology, amplitude, and latency like previous avian studies. Tone-evoked ABRs were found for frequencies from 250 to 4000 Hertz (Hz) and exhibited a range of best sensitivity between 750 and 2000 Hz. Objective click-evoked and tone-evoked ABR thresholds were comparable to subjective thresholds. With these revisited measurements, the chicken ABR still proves to be an excellent example of precocious avian development that complements decades of molecular, neuronal, and systems-level research in the same model organism.
Collapse
Affiliation(s)
- George Ordiway
- Roxelyn and Richard Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, USA
- Central Auditory Physiology Laboratory, Northwestern University, Evanston, IL, USA
| | - Miranda McDonnell
- Roxelyn and Richard Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, USA
- Central Auditory Physiology Laboratory, Northwestern University, Evanston, IL, USA
| | - Jason Tait Sanchez
- Roxelyn and Richard Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, USA
- Central Auditory Physiology Laboratory, Northwestern University, Evanston, IL, USA
- Knowles Hearing Research Center, Northwestern University, Evanston, IL, USA
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| |
Collapse
|
5
|
Wang X, Sela-Donenfeld D, Wang Y. Axonal and presynaptic FMRP: Localization, signal, and functional implications. Hear Res 2023; 430:108720. [PMID: 36809742 PMCID: PMC9998378 DOI: 10.1016/j.heares.2023.108720] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 01/22/2023] [Accepted: 02/09/2023] [Indexed: 02/12/2023]
Abstract
Fragile X mental retardation protein (FMRP) binds a selected set of mRNAs and proteins to guide neural circuit assembly and regulate synaptic plasticity. Loss of FMRP is responsible for Fragile X syndrome, a neuropsychiatric disorder characterized with auditory processing problems and social difficulty. FMRP actions in synaptic formation, maturation, and plasticity are site-specific among the four compartments of a synapse: presynaptic and postsynaptic neurons, astrocytes, and extracellular matrix. This review summarizes advancements in understanding FMRP localization, signals, and functional roles in axons and presynaptic terminals.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University, Guangzhou 510632, China
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Yuan Wang
- Department of Biomedical Sciences, Program in Neuroscience, Florida State University College of Medicine, Tallahassee, FL 32306, USA.
| |
Collapse
|
6
|
Curnow E, Wang Y. New Animal Models for Understanding FMRP Functions and FXS Pathology. Cells 2022; 11:1628. [PMID: 35626665 PMCID: PMC9140010 DOI: 10.3390/cells11101628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/03/2022] [Accepted: 05/09/2022] [Indexed: 11/16/2022] Open
Abstract
Fragile X encompasses a range of genetic conditions, all of which result as a function of changes within the FMR1 gene and abnormal production and/or expression of the FMR1 gene products. Individuals with Fragile X syndrome (FXS), the most common heritable form of intellectual disability, have a full-mutation sequence (>200 CGG repeats) which brings about transcriptional silencing of FMR1 and loss of FMR protein (FMRP). Despite considerable progress in our understanding of FXS, safe, effective, and reliable treatments that either prevent or reduce the severity of the FXS phenotype have not been approved. While current FXS animal models contribute their own unique understanding to the molecular, cellular, physiological, and behavioral deficits associated with FXS, no single animal model is able to fully recreate the FXS phenotype. This review will describe the status and rationale in the development, validation, and utility of three emerging animal model systems for FXS, namely the nonhuman primate (NHP), Mongolian gerbil, and chicken. These developing animal models will provide a sophisticated resource in which the deficits in complex functions of perception, action, and cognition in the human disorder are accurately reflected and aid in the successful translation of novel therapeutics and interventions to the clinic setting.
Collapse
Affiliation(s)
- Eliza Curnow
- REI Division, Department of ObGyn, University of Washington, Seattle, WA 98195, USA
- Washington National Primate Research Center, University of Washington, Seattle, WA 98195, USA
| | - Yuan Wang
- Program in Neuroscience, Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
7
|
Tonotopic Specializations in Number, Size, and Reversal Potential of GABAergic Inputs Fine-Tune Temporal Coding at Avian Cochlear Nucleus. J Neurosci 2021; 41:8904-8916. [PMID: 34518306 DOI: 10.1523/jneurosci.0884-21.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/31/2021] [Accepted: 09/04/2021] [Indexed: 11/21/2022] Open
Abstract
GABAergic inhibition in neurons plays a critical role in determining the output of neural circuits. Neurons in avian nucleus magnocellularis (NM) use several tonotopic-region-dependent specializations to relay the timing information of sound in the auditory nerve to higher auditory nuclei. Previously, we showed that feedforward GABAergic inhibition in NM has a different dependence on the level of auditory nerve activity, with the low-frequency region having a low-threshold and linear relationship, while the high-frequency region has a high-threshold and step-like relationship. However, it remains unclear how the GABAergic synapses are tonotopically regulated and interact with other specializations of NM neurons. In this study, we examined GABAergic transmission in the NM of chickens of both sexes and explored its contributions to the temporal coding of sound at each tonotopic region. We found that the number and size of unitary GABAergic currents and their reversal potential were finely tuned at each tonotopic region in the NM. At the lower-frequency region, unitary GABAergic currents were larger in number but smaller in size. In addition, their reversal potential was close to the resting potential of neurons, which enabled reliable inhibition despite the smaller potassium conductance. At the higher-frequency region, on the other hand, unitary GABAergic currents were fewer, larger, and highly depolarizing, which enabled powerful inhibition via activating the large potassium conductance. Thus, we propose that GABAergic synapses are coordinated with the characteristics of excitatory synapses and postsynaptic neurons, ensuring the temporal coding for wide frequency and intensity ranges.SIGNIFICANCE STATEMENT We found in avian cochlear nucleus that the number and size of unitary GABAergic inputs differed among tonotopic regions and correlated to respective excitatory inputs; it was larger in number but smaller in size for neurons tuned to lower-frequency sound. Furthermore, GABAergic reversal potential also differed among the regions in accordance with the size of Kv1 current; it was less depolarized in the lower-frequency neurons with smaller Kv1 current. These differentiations of GABAergic transmission maximized the effects of inhibition at each tonotopic region, ensuring precise and reliable temporal coding across frequencies and intensities. Our results emphasize the importance of optimizing characteristics of GABAergic transmission within individual neurons for proper neural circuit function.
Collapse
|
8
|
Bondy BJ, Haimes DB, Golding NL. Physiological Diversity Influences Detection of Stimulus Envelope and Fine Structure in Neurons of the Medial Superior Olive. J Neurosci 2021; 41:6234-6245. [PMID: 34083255 PMCID: PMC8287997 DOI: 10.1523/jneurosci.2354-20.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 01/10/2023] Open
Abstract
The neurons of the medial superior olive (MSO) of mammals extract azimuthal information from the delays between sounds reaching the two ears [interaural time differences (ITDs)]. Traditionally, all models of sound localization have assumed that MSO neurons represent a single population of cells with specialized and homogeneous intrinsic and synaptic properties that enable the detection of synaptic coincidence on a timescale of tens to hundreds of microseconds. Here, using patch-clamp recordings from large populations of anatomically labeled neurons in brainstem slices from male and female Mongolian gerbils (Meriones unguiculatus), we show that MSO neurons are far more physiologically diverse than previously appreciated, with properties that depend regionally on cell position along the topographic map of frequency. Despite exhibiting a similar morphology, neurons in the MSO exhibit subthreshold oscillations of differing magnitudes that drive action potentials at rates between 100 and 800 Hz. These oscillations are driven primarily by voltage-gated sodium channels and are distinct from resonant properties derived from other active membrane properties. We show that graded differences in these and other physiological properties across the MSO neuron population enable the MSO to duplex the encoding of ITD information in both fast, submillisecond time-varying signals as well as in slower envelopes.SIGNIFICANCE STATEMENT Neurons in the medial superior olive (MSO) encode sound localization cues by detecting microsecond differences in the arrival times of inputs from the left and right ears, and it has been assumed that this computation is made possible by highly stereotyped structural and physiological specializations. Here we report using a large (>400) sample size in which MSO neurons show a strikingly large continuum of functional properties despite exhibiting similar morphologies. We demonstrate that subthreshold oscillations mediated by voltage-gated Na+ channels play a key role in conferring graded differences in firing properties. This functional diversity likely confers capabilities of processing both fast, submillisecond-scale synaptic activity (acoustic "fine structure"), and slow-rising envelope information that is found in amplitude-modulated sounds and speech patterns.
Collapse
Affiliation(s)
- Brian J Bondy
- Department of Neuroscience, University of Texas at Austin, Austin, Texas 78712
- Center for Learning and Memory, University of Texas at Austin, Austin, Texas 78712
| | - David B Haimes
- Department of Neuroscience, University of Texas at Austin, Austin, Texas 78712
- Center for Learning and Memory, University of Texas at Austin, Austin, Texas 78712
| | - Nace L Golding
- Department of Neuroscience, University of Texas at Austin, Austin, Texas 78712
- Center for Learning and Memory, University of Texas at Austin, Austin, Texas 78712
| |
Collapse
|
9
|
Saleh AJ, Nothwang HG. Differential expression of microRNAs in the developing avian auditory hindbrain. J Comp Neurol 2021; 529:3477-3496. [PMID: 34180540 DOI: 10.1002/cne.25205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 11/08/2022]
Abstract
The avian auditory hindbrain is a longstanding model for studying neural circuit development. Information on gene regulatory network (GRN) components underlying this process, however, is scarce. Recently, the spatiotemporal expression of 12 microRNAs (miRNAs) was investigated in the mammalian auditory hindbrain. As a comparative study, we here investigated the spatiotemporal expression of the orthologous miRNAs during development of the chicken auditory hindbrain. All miRNAs were expressed both at E13, an immature stage, and P14, a mature stage of the auditory system. In most auditory nuclei, a homogeneous expression pattern was observed at both stages, like the mammalian system. An exception was the nucleus magnocellularis (NM). There, at E13, nine miRNAs showed a differential expression pattern along the cochleotopic axis with high expression at the rostromedial pole. One of them showed a gradient expression whereas eight showed a spatially selective expression at the rostral pole that reflected the different rhombomeric origins of this composite nucleus. The miRNA differential expression persisted in the NM to the mature stage, with the selective expression changed to linear gradients. Bioinformatics analysis predicted mRNA targets that are associated with neuronal developmental processes such as neurite and synapse organization, calcium and ephrin-Eph signaling, and neurotransmission. Overall, this first analysis of miRNAs in the chicken central auditory system reveals shared and strikingly distinct features between chicken and murine orthologues. The embryonic gradient expression of these GRN elements in the NM adds miRNA patterns to the list of cochleotopic and developmental gradients in the central auditory system.
Collapse
Affiliation(s)
- Ali Jason Saleh
- Division of Neurogenetics and Cluster of Excellence Hearing4All, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Hans Gerd Nothwang
- Division of Neurogenetics and Cluster of Excellence Hearing4All, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany.,Research Center for Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| |
Collapse
|
10
|
Koert E, Kuenzel T. Small dendritic synapses enhance temporal coding in a model of cochlear nucleus bushy cells. J Neurophysiol 2021; 125:915-937. [PMID: 33471627 DOI: 10.1152/jn.00331.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Spherical bushy cells (SBCs) in the anteroventral cochlear nucleus receive a single or very few powerful axosomatic inputs from the auditory nerve. However, SBCs are also contacted by small regular bouton synapses of the auditory nerve, located in their dendritic tree. The function of these small inputs is unknown. It was speculated that the interaction of axosomatic inputs with small dendritic inputs improved temporal precision, but direct evidence for this is missing. In a compartment model of spherical bushy cells with a stylized or realistic three-dimensional (3-D) representation of the bushy dendrite, we explored this hypothesis. Phase-locked dendritic inputs caused both tonic depolarization and a modulation of the model SBC membrane potential at the frequency of the stimulus. For plausible model parameters, dendritic inputs were subthreshold. Instead, the tonic depolarization increased the excitability of the SBC model and the modulation of the membrane potential caused a phase-dependent increase in the efficacy of the main axosomatic input. This improved response rate and entrainment for low-input frequencies and temporal precision of output at and above the characteristic frequency. A careful exploration of morphological and biophysical parameters of the bushy dendrite suggested a functional explanation for the peculiar shape of the bushy dendrite. Our model for the first time directly implied a role for the small excitatory dendritic inputs in auditory processing: they modulate the efficacy of the main input and are thus a plausible mechanism for the improvement of temporal precision and fidelity in these central auditory neurons.NEW & NOTEWORTHY We modeled dendritic inputs from the auditory nerve that spherical bushy cells of the cochlear nucleus receive. Dendritic inputs caused both tonic depolarization and modulation of the membrane potential at the input frequency. This improved the rate, entrainment, and temporal precision of output action potentials. Our simulations suggest a role for small dendritic inputs in auditory processing: they modulate the efficacy of the main input supporting temporal precision and fidelity in these central auditory neurons.
Collapse
Affiliation(s)
- Elisabeth Koert
- Auditory Neurophysiology Group, Department of Chemosensation, RWTH Aachen University, Aachen, Germany
| | - Thomas Kuenzel
- Auditory Neurophysiology Group, Department of Chemosensation, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
11
|
Takahashi M, Sanchez JT. Effects of Neurotrophin-3 on Intrinsic Neuronal Properties at a Central Auditory Structure. Neurosci Insights 2020; 15:2633105520980442. [PMID: 33354669 PMCID: PMC7734498 DOI: 10.1177/2633105520980442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 11/23/2020] [Indexed: 11/15/2022] Open
Abstract
Neurotrophins, a class of growth factor proteins that control neuronal proliferation, morphology, and apoptosis, are found ubiquitously throughout the nervous system. One particular neurotrophin (NT-3) and its cognate tyrosine receptor kinase (TrkC) have recently received attention as a possible therapeutic target for synaptopathic sensorineural hearing loss. Additionally, research shows that NT-3-TrkC signaling plays a role in establishing the sensory organization of frequency topology (ie, tonotopic order) in the cochlea of the peripheral inner ear. However, the neurotrophic effects of NT-3 on central auditory properties are unclear. In this study we examined whether NT-3-TrkC signaling affects the intrinsic electrophysiological properties at a first-order central auditory structure in chicken, known as nucleus magnocellularis (NM). Here, the expression pattern of specific neurotrophins is well known and tightly regulated. By using whole-cell patch-clamp electrophysiology, we show that NT-3 application to brainstem slices does not affect intrinsic properties of high-frequency neuronal regions but had robust effects for low-frequency neurons, altering voltage-dependent potassium functions, action potential repolarization kinetics, and passive membrane properties. We suggest that NT-3 may contribute to the precise establishment and organization of tonotopy in the central auditory pathway by playing a specialized role in regulating the development of intrinsic neuronal properties of low-frequency NM neurons.
Collapse
Affiliation(s)
- Momoko Takahashi
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, USA
| | - Jason Tait Sanchez
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, USA
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
- The Hugh Knowles Hearing Research Center, Northwestern University, Evanston, IL, USA
| |
Collapse
|
12
|
Wang X, Kohl A, Yu X, Zorio DAR, Klar A, Sela-Donenfeld D, Wang Y. Temporal-specific roles of fragile X mental retardation protein in the development of the hindbrain auditory circuit. Development 2020; 147:dev.188797. [PMID: 32747436 DOI: 10.1242/dev.188797] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 06/29/2020] [Indexed: 01/01/2023]
Abstract
Fragile X mental retardation protein (FMRP) is an RNA-binding protein abundant in the nervous system. Functional loss of FMRP leads to sensory dysfunction and severe intellectual disabilities. In the auditory system, FMRP deficiency alters neuronal function and synaptic connectivity and results in perturbed processing of sound information. Nevertheless, roles of FMRP in embryonic development of the auditory hindbrain have not been identified. Here, we developed high-specificity approaches to genetically track and manipulate throughout development of the Atoh1+ neuronal cell type, which is highly conserved in vertebrates, in the cochlear nucleus of chicken embryos. We identified distinct FMRP-containing granules in the growing axons of Atoh1+ neurons and post-migrating NM cells. FMRP downregulation induced by CRISPR/Cas9 and shRNA techniques resulted in perturbed axonal pathfinding, delay in midline crossing, excess branching of neurites, and axonal targeting errors during the period of circuit development. Together, these results provide the first in vivo identification of FMRP localization and actions in developing axons of auditory neurons, and demonstrate the importance of investigating early embryonic alterations toward understanding the pathogenesis of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Department of Biomedical Sciences, Program in Neuroscience, Florida State University College of Medicine, Tallahassee, FL 32306, USA.,Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University, Guangzhou 510632, China
| | - Ayelet Kohl
- Koret School of Veterinary Medicine, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Xiaoyan Yu
- Department of Biomedical Sciences, Program in Neuroscience, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Diego A R Zorio
- Department of Biomedical Sciences, Program in Neuroscience, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - Avihu Klar
- Department of Medical Neurobiology IMRIC, Hebrew University Medical School, Jerusalem 91120, Israel
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Yuan Wang
- Department of Biomedical Sciences, Program in Neuroscience, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| |
Collapse
|
13
|
Structural and Functional Refinement of the Axon Initial Segment in Avian Cochlear Nucleus during Development. J Neurosci 2020; 40:6709-6721. [PMID: 32719016 DOI: 10.1523/jneurosci.3068-19.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 05/25/2020] [Accepted: 06/26/2020] [Indexed: 12/12/2022] Open
Abstract
The axon initial segment (AIS) is involved in action potential initiation. Structural and biophysical characteristics of the AIS differ among cell types and/or brain regions, but the underlying mechanisms remain elusive. Using immunofluorescence and electrophysiological methods, combined with super-resolution imaging, we show in the developing nucleus magnocellularis of the chicken in both sexes that the AIS is refined in a tonotopic region-dependent manner. This process of AIS refinement differs among cells tuned to different frequencies. At hearing onset, the AIS was ∼50 µm long with few voltage-gated sodium channels regardless of tonotopic region. However, after hatching, the AIS matured and displayed an ∼20-µm-long structure with a significant enrichment of sodium channels responsible for an increase in sodium current and a decrease in spike threshold. Moreover, the shortening was more pronounced, while the accumulation of channels was not, in neurons tuned to higher frequency, creating tonotopic differences in the AIS. We conclude that AIS shortening is mediated by disassembly of the cytoskeleton at the distal end of the AIS, despite intact periodicity of the submembranous cytoskeleton across the AIS. Importantly, deprivation of afferent input diminished the shortening in neurons tuned to a higher frequency to a larger extent in posthatch animals, with little effect on the accumulation of sodium channels. Thus, cytoskeletal reorganization and sodium channel enrichment at the AIS are differentially regulated depending on tonotopic region, but work synergistically to optimize neuronal output in the auditory nucleus.SIGNIFICANCE STATEMENT The axon initial segment (AIS) plays fundamental roles in determining neuronal output. The AIS varies structurally and molecularly across tonotopic regions in avian cochlear nucleus. However, the mechanism underlying these variations remains unclear. The AIS is immature around hearing onset, but becomes shorter and accumulates more sodium channels during maturation, with a pronounced shortening and a moderate channel accumulation at higher tonotopic regions. Afferent input adjusts sodium conductance at the AIS by augmenting AIS shortening (via disassembly of cytoskeletons at its distal end) specifically at higher-frequency regions. However, this had little effect on channel accumulation. Thus, cytoskeletal structure and sodium channel accumulation at the AIS are regulated differentially but work synergistically to optimize the neuronal output.
Collapse
|
14
|
Yu X, Wang X, Sakano H, Zorio DAR, Wang Y. Dynamics of the fragile X mental retardation protein correlates with cellular and synaptic properties in primary auditory neurons following afferent deprivation. J Comp Neurol 2020; 529:481-500. [PMID: 32449186 DOI: 10.1002/cne.24959] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/26/2020] [Accepted: 05/14/2020] [Indexed: 01/01/2023]
Abstract
Afferent activity dynamically regulates neuronal properties and connectivity in the central nervous system. The Fragile X mental retardation protein (FMRP) is an RNA-binding protein that regulates cellular and synaptic properties in an activity-dependent manner. Whether and how FMRP level and localization are regulated by afferent input remains sparsely examined and how such regulation is associated with neuronal response to changes in sensory input is unknown. We characterized changes in FMRP level and localization in the chicken nucleus magnocellularis (NM), a primary cochlear nucleus, following afferent deprivation by unilateral cochlea removal. We observed rapid (within 2 hr) aggregation of FMRP immunoreactivity into large granular structures in a subset of deafferented NM neurons. Neurons that exhibited persistent FMRP aggregation at 12-24 hr eventually lost cytoplasmic Nissl substance, indicating cell death. A week later, FMRP expression in surviving neurons regained its homeostasis, with a slightly reduced immunostaining intensity and enhanced heterogeneity. Correlation analyses under the homeostatic status (7-14 days) revealed that neurons expressing relatively more FMRP had a higher capability of maintaining cell body size and ribosomal activity, as well as a better ability to detach inactive presynaptic terminals. Additionally, the intensity of an inhibitory postsynaptic protein, gephyrin, was reduced following deafferentation and was positively correlated with FMRP intensity, implicating an involvement of FMRP in synaptic dynamics in response to reduced afferent inputs. Collectively, this study demonstrates that afferent input regulates FMRP expression and localization in ways associated with multiple types of neuronal responses and synaptic rearrangements.
Collapse
Affiliation(s)
- Xiaoyan Yu
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, USA
| | - Xiaoyu Wang
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, USA.,Division of Histology & Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, Medical College, Jinan University, Guangzhou, China
| | - Hitomi Sakano
- Department of Otolaryngology, Bloedel Hearing Research Center, University of Washington, Seattle, Washington, USA.,Department of Otolaryngology, University of Rochester, Rochester, New York, USA
| | - Diego A R Zorio
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, USA
| | - Yuan Wang
- Program in Neuroscience, Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida, USA
| |
Collapse
|
15
|
Adachi R, Yamada R, Kuba H. Tonotopic Differentiation of Coupling between Ca 2+ and Kv1.1 Expression in Brainstem Auditory Circuit. iScience 2019; 13:199-213. [PMID: 30856389 PMCID: PMC6411580 DOI: 10.1016/j.isci.2019.02.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 12/17/2018] [Accepted: 02/22/2019] [Indexed: 12/25/2022] Open
Abstract
Tonotopic differentiations of ion channels ensure sound processing across frequencies. Afferent input plays a critical role in differentiations. We demonstrate here in organotypic culture of chicken cochlear nucleus that expression of Kv1.1 was coupled with Ca2+ to a different degree depending on tonotopic regions, thereby differentiating the level of expression within the nucleus. In the culture, Kv1.1 was down-regulated and not differentiated tonotopically. Chronic depolarization increased Kv1.1 expression in a level-dependent manner. Moreover, the dependence was steeper at higher-frequency regions, which restored the differentiation. The depolarization increased Kv1.1 via activation of Cav1 channels, whereas basal Ca2+ level elevated similarly irrespective of tonotopic regions. Thus, the efficiency of Ca2+-dependent Kv1.1 expression would be fine-tuned in a tonotopic-region-specific manner, emphasizing the importance of neuronal tonotopic identity as well as pattern of afferent input in the tonotopic differentiation of the channel in the auditory circuit. Kv1.1 expression is down-regulated in slice culture of chicken cochlear nucleus Depolarization up-regulates Kv1.1 in a tonotopic-region-specific manner Level of Kv1.1 expression is dependent on basal calcium concentration Efficiency of calcium-dependent Kv1.1 expression is differentiated tonotopically
Collapse
Affiliation(s)
- Ryota Adachi
- Department of Cell Physiology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Rei Yamada
- Department of Cell Physiology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan
| | - Hiroshi Kuba
- Department of Cell Physiology, Graduate School of Medicine, Nagoya University, Nagoya 466-8550, Japan.
| |
Collapse
|
16
|
Brown DH, Hyson RL. Intrinsic physiological properties underlie auditory response diversity in the avian cochlear nucleus. J Neurophysiol 2019; 121:908-927. [PMID: 30649984 DOI: 10.1152/jn.00459.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Sensory systems exploit parallel processing of stimulus features to enable rapid, simultaneous extraction of information. Mechanisms that facilitate this differential extraction of stimulus features can be intrinsic or synaptic in origin. A subdivision of the avian cochlear nucleus, nucleus angularis (NA), extracts sound intensity information from the auditory nerve and contains neurons that exhibit diverse responses to sound and current injection. NA neurons project to multiple regions ascending the auditory brain stem including the superior olivary nucleus, lateral lemniscus, and avian inferior colliculus, with functional implications for inhibitory gain control and sound localization. Here we investigated whether the diversity of auditory response patterns in NA can be accounted for by variation in intrinsic physiological features. Modeled sound-evoked auditory nerve input was applied to NA neurons with dynamic clamp during in vitro whole cell recording at room temperature. Temporal responses to auditory nerve input depended on variation in intrinsic properties, and the low-threshold K+ current was implicated as a major contributor to temporal response diversity and neuronal input-output functions. An auditory nerve model of acoustic amplitude modulation produced synchrony coding of modulation frequency that depended on the intrinsic physiology of the individual neuron. In Primary-Like neurons, varying low-threshold K+ conductance with dynamic clamp altered temporal modulation tuning bidirectionally. Taken together, these data suggest that intrinsic physiological properties play a key role in shaping auditory response diversity to both simple and more naturalistic auditory stimuli in the avian cochlear nucleus. NEW & NOTEWORTHY This article addresses the question of how the nervous system extracts different information in sounds. Neurons in the cochlear nucleus show diverse responses to acoustic stimuli that may allow for parallel processing of acoustic features. The present studies suggest that diversity in intrinsic physiological features of individual neurons, including levels of a low voltage-activated K+ current, play a major role in regulating the diversity of auditory responses.
Collapse
Affiliation(s)
- David H Brown
- Program in Neuroscience, Department of Psychology, Florida State University , Tallahassee, Florida
| | - Richard L Hyson
- Program in Neuroscience, Department of Psychology, Florida State University , Tallahassee, Florida
| |
Collapse
|
17
|
Hong H, Sanchez JT. Need for Speed and Precision: Structural and Functional Specialization in the Cochlear Nucleus of the Avian Auditory System. J Exp Neurosci 2018; 12:1179069518815628. [PMID: 30559595 PMCID: PMC6291874 DOI: 10.1177/1179069518815628] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/06/2018] [Indexed: 11/17/2022] Open
Abstract
Birds such as the barn owl and zebra finch are known for their remarkable hearing abilities that are critical for survival, communication, and vocal learning functions. A key to achieving these hearing abilities is the speed and precision required for the temporal coding of sound-a process heavily dependent on the structural, synaptic, and intrinsic specializations in the avian auditory brainstem. Here, we review recent work from us and others focusing on the specialization of neurons in the chicken cochlear nucleus magnocellularis (NM)-a first-order auditory brainstem structure analogous to bushy cells in the mammalian anteroventral cochlear nucleus. Similar to their mammalian counterpart, NM neurons are mostly adendritic and receive auditory nerve input through large axosomatic endbulb of Held synapses. Axonal projections from NM neurons to their downstream auditory targets are sophisticatedly programmed regarding their length, caliber, myelination, and conduction velocity. Specialized voltage-dependent potassium and sodium channel properties also play important and unique roles in shaping the functional phenotype of NM neurons. Working synergistically with potassium channels, an atypical current known as resurgent sodium current promotes rapid and precise action potential firing for NM neurons. Interestingly, these structural and functional specializations vary dramatically along the tonotopic axis and suggest a plethora of encoding strategies for sounds of different acoustic frequencies, mechanisms likely shared across species.
Collapse
Affiliation(s)
- Hui Hong
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, USA
| | - Jason Tait Sanchez
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, USA.,Department of Neurobiology, Northwestern University, Evanston, IL, USA.,The Hugh Knowles Hearing Research Center, Northwestern University, Evanston, IL, USA
| |
Collapse
|
18
|
Wang X, Zorio DAR, Schecterson L, Lu Y, Wang Y. Postsynaptic FMRP Regulates Synaptogenesis In Vivo in the Developing Cochlear Nucleus. J Neurosci 2018; 38:6445-6460. [PMID: 29950504 PMCID: PMC6052239 DOI: 10.1523/jneurosci.0665-18.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/18/2018] [Accepted: 06/20/2018] [Indexed: 12/29/2022] Open
Abstract
A global loss of the fragile X mental retardation protein (FMRP; encoded by the Fmr1 gene) leads to sensory dysfunction and intellectual disabilities. One underlying mechanism of these phenotypes is structural and functional deficits in synapses. Here, we determined the autonomous function of postsynaptic FMRP in circuit formation, synaptogenesis, and synaptic maturation. In normal cochlea nucleus, presynaptic auditory axons form large axosomatic endbulb synapses on cell bodies of postsynaptic bushy neurons. In ovo electroporation of drug-inducible Fmr1-shRNA constructs produced a mosaicism of FMRP expression in chicken (either sex) bushy neurons, leading to reduced FMRP levels in transfected, but not neighboring nontransfected, neurons. Structural analyses revealed that postsynaptic FMRP reduction led to smaller size and abnormal morphology of individual presynaptic endbulbs at both early and later developmental stages. We further examined whether FMRP reduction affects dendritic development, as a potential mechanism underlying defective endbulb formation. Normally, chicken bushy neurons grow extensive dendrites at early stages and retract these dendrites when endbulbs begin to form. Neurons transfected with Fmr1 shRNA exhibited a remarkable delay in branch retraction, failing to provide necessary somatic surface for timely formation and growth of large endbulbs. Patch-clamp recording verified functional consequences of dendritic and synaptic deficits on neurotransmission, showing smaller amplitudes and slower kinetics of spontaneous and evoked EPSCs. Together, these data demonstrate that proper levels of postsynaptic FMRP are required for timely maturation of somatodendritic morphology, a delay of which may affect synaptogenesis and thus contribute to long-lasting deficits of excitatory synapses.SIGNIFICANCE STATEMENT Fragile X mental retardation protein (FMRP) regulates a large variety of neuronal activities. A global loss of FMRP affects neural circuit development and synaptic function, leading to fragile X syndrome (FXS). Using temporally and spatially controlled genetic manipulations, this study provides the first in vivo report that autonomous FMRP regulates multiple stages of dendritic development, and that selective reduction of postsynaptic FMRP leads to abnormal development of excitatory presynaptic terminals and compromised neurotransmission. These observations demonstrate secondary influence of developmentally transient deficits in neuronal morphology and connectivity to the development of long-lasting synaptic pathology in FXS.
Collapse
Affiliation(s)
- Xiaoyu Wang
- Department of Biomedical Science, Program in Neuroscience, Florida State University College of Medicine, Tallahassee, Florida 32306
| | - Diego A R Zorio
- Department of Biomedical Science, Program in Neuroscience, Florida State University College of Medicine, Tallahassee, Florida 32306
| | - Leslayann Schecterson
- Department of Otolaryngology, Bloedel Hearing Research Center, University of Washington, Seattle, Washington 98195, and
| | - Yong Lu
- Department of Anatomy and Neurobiology, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio 44272
| | - Yuan Wang
- Department of Biomedical Science, Program in Neuroscience, Florida State University College of Medicine, Tallahassee, Florida 32306,
| |
Collapse
|
19
|
Hong H, Wang X, Lu T, Zorio DAR, Wang Y, Sanchez JT. Diverse Intrinsic Properties Shape Functional Phenotype of Low-Frequency Neurons in the Auditory Brainstem. Front Cell Neurosci 2018; 12:175. [PMID: 29997479 PMCID: PMC6028565 DOI: 10.3389/fncel.2018.00175] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 06/04/2018] [Indexed: 12/18/2022] Open
Abstract
In the auditory system, tonotopy is the spatial arrangement of where sounds of different frequencies are processed. Defined by the organization of neurons and their inputs, tonotopy emphasizes distinctions in neuronal structure and function across topographic gradients and is a common feature shared among vertebrates. In this study we characterized action potential firing patterns and ion channel properties from neurons located in the extremely low-frequency region of the chicken nucleus magnocellularis (NM), an auditory brainstem structure. We found that NM neurons responsible for encoding the lowest sound frequencies (termed NMc neurons) have enhanced excitability and fired bursts of action potentials to sinusoidal inputs ≤10 Hz; a distinct firing pattern compared to higher-frequency neurons. This response property was due to lower amounts of voltage dependent potassium (KV) conductances, unique combination of KV subunits and specialized sodium (NaV) channel properties. Particularly, NMc neurons had significantly lower KV1 and KV3 currents, but higher KV2 current. NMc neurons also showed larger and faster transient NaV current (INaT) with different voltage dependence of inactivation from higher-frequency neurons. In contrast, significantly smaller resurgent sodium current (INaR) was present in NMc with kinetics and voltage dependence that differed from higher-frequency neurons. Immunohistochemistry showed expression of NaV1.6 channel subtypes across the tonotopic axis. However, various immunoreactive patterns were observed between regions, likely underlying some tonotopic differences in INaT and INaR. Finally, using pharmacology and computational modeling, we concluded that KV3, KV2 channels and INaR work synergistically to regulate burst firing in NMc.
Collapse
Affiliation(s)
- Hui Hong
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, United States
| | - Xiaoyu Wang
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, United States
- Program in Neuroscience Florida State University College of Medicine, Florida State University, Tallahassee, FL, United States
| | - Ting Lu
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, United States
| | - Diego A. R. Zorio
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, United States
- Program in Neuroscience Florida State University College of Medicine, Florida State University, Tallahassee, FL, United States
| | - Yuan Wang
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, United States
- Program in Neuroscience Florida State University College of Medicine, Florida State University, Tallahassee, FL, United States
| | - Jason Tait Sanchez
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, United States
- Department of Neurobiology, Northwestern University, Evanston, IL, United States
- The Hugh Knowles Hearing Research Center, Northwestern University, Evanston, IL, United States
| |
Collapse
|
20
|
Auditory Input Shapes Tonotopic Differentiation of Kv1.1 Expression in Avian Cochlear Nucleus during Late Development. J Neurosci 2018; 38:2967-2980. [PMID: 29439165 DOI: 10.1523/jneurosci.2472-17.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 01/30/2018] [Accepted: 02/05/2018] [Indexed: 11/21/2022] Open
Abstract
Tonotopic differentiation is fundamental for signal processing in the auditory system. However, when and how this differentiation arises remain elusive. We addressed this issue using electrophysiology and immunohistochemistry in nucleus magnocellularis of chickens of both sexes, which is known to differ in the expression of Kv1.1 channels depending on characteristic frequency (CF). Just after hearing onset (embryonic day 12-14), Kv1 current gradually increased to a slightly larger extent in neurons with higher CF, causing a tonotopic difference of Kv1 current before hatch. However, after hatch, a much larger increase of Kv1 current occurred, particularly in higher-CF neurons, due to an augmentation of Kv1.1 expression at the plasma membrane. This later change in expression led to the large tonotopic difference of Kv1 current characteristic of mature animals. Attenuation of auditory input by inducing conductive or sensorineural hearing loss around hatch suppressed the differentiation in a level-dependent manner. Moreover, elevation of auditory input during embryonic periods could not reproduce the differentiation, suggesting that the capacity of neurons to drive Kv1.1 expression via auditory input develops in a cell-specific manner, thus underlying the frequency-specific expression of the channel within the nucleus. The results indicated that the tonotopic differentiation of Kv1.1 in nucleus magnocellularis is partially determined before hatch, but largely driven by afferent input after hatch. Our results highlight the importance of neuronal capacity for sound to drive ion channel expression as well as the level of auditory experience in the frequency tuning of brainstem auditory circuits.SIGNIFICANCE STATEMENT Tuning-frequency-specific expression of ion channels is a prerequisite for auditory system function, but its underlying mechanisms remain unclear. Here, we revealed in avian cochlear nucleus that the expression of Kv1.1 became more dependent on auditory input at a late period of maturation in neurons tuned to higher-frequency sound, leading to frequency-specific Kv1.1 expression. Attenuation of auditory input during this period suppressed the differentiation in a level-dependent manner, whereas elevation of input in earlier periods could not reproduce the differentiation. Thus, the capacity of neurons to drive Kv1.1 expression via auditory input develops in a cell-specific manner and directs differentiation, highlighting the importance of neuronal character as well as the level of input in the frequency tuning of auditory circuits.
Collapse
|