1
|
Lorincz D, Drury HR, Lim R, Brichta AM. Immunohistochemical Identification of Sensory Neuropeptides Calcitonin Gene-Related Peptide, Substance P, and Pituitary Adenylate Cyclase-Activating Polypeptide in Efferent Vestibular Nucleus Neurons. Neuroendocrinology 2024; 115:269-282. [PMID: 39662068 PMCID: PMC11991750 DOI: 10.1159/000542984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024]
Abstract
INTRODUCTION The efferent vestibular system (EVS) originates in brainstem efferent vestibular nuclei (EVN) and modifies afferent vestibular signals at their source, in peripheral vestibular organs. Recent evidence suggests that EVS is also involved in the development of motion sickness symptoms, including vertigo and nausea, but the underlying mechanism is unknown. One possible link between EVN and motion sickness symptoms is through the neuropeptide calcitonin gene-related peptide (CGRP). CGRP often co-exists with substance P and pituitary adenylate cyclase-activating polypeptide (PACAP), two neuropeptides with similar vasodilatory effects. Collectively, these sensory neuropeptides have been associated with vestibular migraine pathophysiology and motion sickness. While CGRP and the fast EVS neurotransmitter, acetylcholine (ACh), have previously been identified in EVN neurons and their peripheral terminals, the presence of substance P and PACAP in the EVN has not yet been described. METHODS We used fluorescent immunohistochemistry combined with confocal microscopy to examine the distribution of these three neuropeptides in the mouse EVN. In transgenic choline acetyltransferase (ChAT)-gCaMP6f mice, EVN neurons were positively identified using the fluorescent expression of gCaMP6f. In wild-type C57/BL6 mice, EVN neurons were confirmed using ChAT immunolabelling. RESULTS Consistent with previous studies, CGRP was labelled in a subset of cholinergic EVN neurons. Additionally, we also show evidence for substance P and PACAP expression in EVN of transgenic and wild-type mice. CONCLUSION The presence of CGRP, substance P, and PACAP in EVN neurons suggests a complex peptidergic modulation of cholinergic signalling, whose release into local blood vessels may contribute to motion sickness symptoms.
Collapse
Affiliation(s)
- David Lorincz
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
- Children’s Cancer Institute Australia, Lowy Cancer Research Centre, UNSW Sydney, Sydney, NSW, Australia
- UNSW Medicine and Health, UNSW, Sydney, NSW, Australia
| | - Hannah Rose Drury
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Rebecca Lim
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Alan Martin Brichta
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
2
|
Mohamed NMM, Meredith FL, Rennie KJ. Inhibition of Ionic Currents by Fluoxetine in Vestibular Calyces in Different Epithelial Loci. Int J Mol Sci 2024; 25:8801. [PMID: 39201487 PMCID: PMC11354711 DOI: 10.3390/ijms25168801] [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: 06/27/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
Previous studies have suggested a role for selective serotonin reuptake inhibitors (SSRIs) such as fluoxetine (Prozac®) in the treatment of dizziness and inner ear vestibular dysfunction. The potential mechanism of action within the vestibular system remains unclear; however, fluoxetine has been reported to block certain types of K+ channel in other systems. Here, we investigated the direct actions of fluoxetine on membrane currents in presynaptic hair cells and postsynaptic calyx afferents of the gerbil peripheral vestibular system using whole cell patch clamp recordings in crista slices. We explored differences in K+ currents in peripheral zone (PZ) and central zone (CZ) calyces of the crista and their response to fluoxetine application. Outward K+ currents in PZ calyces showed greater inactivation at depolarized membrane potentials compared to CZ calyces. The application of 100 μM fluoxetine notably reduced K+ currents in calyx terminals within both zones of the crista, and the remaining currents exhibited distinct traits. In PZ cells, fluoxetine inhibited a non-inactivating K+ current and revealed a rapidly activating and inactivating K+ current, which was sensitive to blocking by 4-aminopyridine. This was in contrast to CZ calyces, where low-voltage-activated and non-inactivating K+ currents persisted following application of 100 μM fluoxetine. Additionally, marked inhibition of transient inward Na+ currents by fluoxetine was observed in calyces from both crista zones. Different concentrations of fluoxetine were tested, and the EC50 values were found to be 40 µM and 32 µM for K+ and Na+ currents, respectively. In contrast, 100 μM fluoxetine had no impact on voltage-dependent K+ currents in mechanosensory type I and type II vestibular hair cells. In summary, micromolar concentrations of fluoxetine are expected to strongly reduce both Na+ and K+ conductance in afferent neurons of the peripheral vestibular system in vivo. This would lead to inhibition of action potential firing in vestibular sensory neurons and has therapeutic implications for disorders of balance.
Collapse
Affiliation(s)
| | | | - Katherine J. Rennie
- Department of Otolaryngology-Head & Neck Surgery, University of Colorado School of Medicine, Aurora, CO 80045, USA; (N.M.M.M.); (F.L.M.)
| |
Collapse
|
3
|
Sinha AK, Lee C, Holt JC. KCNQ2/3 regulates efferent mediated slow excitation of vestibular afferents in mammals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.30.573731. [PMID: 38260489 PMCID: PMC10802244 DOI: 10.1101/2023.12.30.573731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Primary vestibular afferents transmit information from hair cells about head position and movement to the CNS, which is critical for maintaining balance, gaze stability and spatial navigation. The CNS, in turn, modulates hair cells and afferents via the efferent vestibular system (EVS) and its activation of several cholinergic signaling mechanisms. Electrical stimulation of EVS neurons gives rise to three kinetically- and mechanistically-distinct afferent responses including a slow excitation, a fast excitation, and a fast inhibition. EVS-mediated slow excitation is attributed to odd-numbered muscarinic acetylcholine receptors (mAChRs) on the afferent whose activation leads to the closure of a potassium conductance and increased afferent discharge. Likely effector candidates include low-threshold, voltage-gated potassium channels belonging to the KCNQ (Kv7.X) family, which are involved in neuronal excitability across the nervous system and are subject to mAChR modulation. Specifically, KCNQ2/3 heteromeric channels may be the molecular correlates for the M-current, a potassium current that is blocked following the activation of odd-numbered mAChRs. To this end, multiple members of the KCNQ channel family, including KCNQ2 and KCNQ3, are localized to several microdomains within vestibular afferent endings, where they influence afferent excitability and could be targeted by EVS neurons. Additionally, the relative expression of KCNQ subunits appears to vary across the sensory epithelia and among different afferent types. However, it is unclear which KCNQ channel subunits are targeted by mAChR activation and whether that also varies among different afferent classes. Here we show that EVS-mediated slow excitation is blocked and enhanced by the non-selective KCNQ channel blocker XE991 and opener retigabine, respectively. Using KCNQ subunit-selective drugs, we observed that a KCNQ2 blocker blocks the slow response in irregular afferents, while a KCNQ2/3 opener enhances slow responses in regular afferents. The KCNQ2 blockers did not appear to affect resting afferent discharge rates, while KCNQ2/3 or KCNQ2/4 openers decreased afferent excitability. Here, we show pharmacological evidence that KCNQ2/3 subunits are likely targeted by mAChR activation in mammalian vestibular afferents. Additionally, we show that KCNQ3 KO mice have altered resting discharge rate as well as EVS-mediated slow response. These data together suggest that KCNQ channels play a role in slow response and discharge rate of vestibular afferents, which can be modulated by EVS in mammals.
Collapse
|
4
|
Sinha AK, Lee C, Holt JC. Elucidating the role of muscarinic acetylcholine receptor (mAChR) signaling in efferent mediated responses of vestibular afferents in mammals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.31.549902. [PMID: 37577578 PMCID: PMC10418111 DOI: 10.1101/2023.07.31.549902] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
The peripheral vestibular system detects head position and movement through activation of vestibular hair cells (HCs) in vestibular end organs. HCs transmit this information to the CNS by way of primary vestibular afferent neurons. The CNS, in turn, modulates HCs and afferents via the efferent vestibular system (EVS) through activation of cholinergic signaling mechanisms. In mice, we previously demonstrated that activation of muscarinic acetylcholine receptors (mAChRs), during EVS stimulation, gives rise to a slow excitation that takes seconds to peak and tens of seconds to decay back to baseline. This slow excitation is mimicked by muscarine and ablated by the non-selective mAChR blockers scopolamine, atropine, and glycopyrrolate. While five distinct mAChRs (M1-M5) exist, the subtype(s) driving EVS-mediated slow excitation remain unidentified and details on how these mAChRs alter vestibular function is not well understood. The objective of this study is to characterize which mAChR subtypes drive the EVS-mediated slow excitation, and how their activation impacts vestibular physiology and behavior. In C57Bl/6J mice, M3mAChR antagonists were more potent at blocking slow excitation than M1mAChR antagonists, while M2/M4 blockers were ineffective. While unchanged in M2/M4mAChR double KO mice, EVS-mediated slow excitation in M3 mAChR-KO animals were reduced or absent in irregular afferents but appeared unchanged in regular afferents. In agreement, vestibular sensory-evoked potentials (VsEP), known to be predominantly generated from irregular afferents, were significantly less enhanced by mAChR activation in M3mAChR-KO mice compared to controls. Finally, M3mAChR-KO mice display distinct behavioral phenotypes in open field activity, and thermal profiles, and balance beam and forced swim test. M3mAChRs mediate efferent-mediated slow excitation in irregular afferents, while M1mAChRs may drive the same process in regular afferents.
Collapse
|
5
|
Bronson D, Kalluri R. Muscarinic Acetylcholine Receptors Modulate HCN Channel Properties in Vestibular Ganglion Neurons. J Neurosci 2023; 43:902-917. [PMID: 36604171 PMCID: PMC9908319 DOI: 10.1523/jneurosci.2552-21.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 12/14/2022] [Accepted: 12/22/2022] [Indexed: 01/07/2023] Open
Abstract
Efferent modulation of vestibular afferent excitability is linked to muscarinic signaling cascades that close low-voltage-gated potassium channels (i.e., KCNQ). Here, we show that muscarinic signaling cascades also depolarize the activation range of hyperpolarization-activated cyclic-nucleotide gated (HCN) channels. We compared the voltage activation range and kinetics of HCN channels and induced firing patterns before and after administering the muscarinic acetylcholine receptor (mAChR) agonist oxotremorine-M (Oxo-M) in dissociated vestibular ganglion neurons (VGNs) from rats of either sex using perforated whole-cell patch-clamp methods. Oxo-M depolarized HCN channels' half-activation voltage (V 1/2) and sped up the rate of activation near resting potential twofold. HCN channels in large-diameter and/or transient firing VGN (putative cell bodies of irregular firing neuron from central epithelial zones) had relatively depolarized V 1/2 in control solution and were less sensitive to mAChR activation than those found in small-diameter VGN with sustained firing patterns (putatively belonging to regular firing afferents). The impact of mAChR on HCN channels is not a direct consequence of closing KCNQ channels since pretreating the cells with Linopirdine, a KCNQ channel blocker, did not prevent HCN channel depolarization by Oxo-M. Efferent signaling promoted ion channel configurations that were favorable to highly regular spiking in some VGN, but not others. This is consistent with previous observations that low-voltage gated potassium currents in VGN are conducted by mAChR agonist-sensitive and -insensitive channels. Connecting efferent signaling to HCN channels is significant because of the channel's impact on spike-timing regularity and nonchemical transmission between Type I hair cells and vestibular afferents.SIGNIFICANCE STATEMENT Vestibular afferents express a diverse complement of ion channels. In vitro studies identified low-voltage activated potassium channels and hyperpolarization-activated cyclic-nucleotide gated (HCN) channels as crucial for shaping the timing and sensitivity of afferent responses. Moreover, a network of acetylcholine-releasing efferent neurons controls afferent excitability by closing a subgroup of low-voltage activated potassium channels on the afferent neuron. This work shows that these efferent signaling cascades also enhance the activation of HCN channels by depolarizing their voltage activation range. The size of this effect varies depending on the endogenous properties of the HCN channel and on cell type (as determined by discharge patterns and cell size). Simultaneously controlling two ion-channel groups gives the vestibular efferent system exquisite control over afferent neuron activity.
Collapse
Affiliation(s)
- Daniel Bronson
- Hearing and Communications Neuroscience Training Program, University of Southern California, Los Angeles, California 90057
- Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California 90057
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California 90057
| | - Radha Kalluri
- Hearing and Communications Neuroscience Training Program, University of Southern California, Los Angeles, California 90057
- Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California 90057
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California 90057
| |
Collapse
|
6
|
Govindaraju AC, Quraishi IH, Lysakowski A, Eatock RA, Raphael RM. Nonquantal transmission at the vestibular hair cell-calyx synapse: K LV currents modulate fast electrical and slow K + potentials. Proc Natl Acad Sci U S A 2023; 120:e2207466120. [PMID: 36595693 PMCID: PMC9926171 DOI: 10.1073/pnas.2207466120] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Vestibular hair cells transmit information about head position and motion across synapses to primary afferent neurons. At some of these synapses, the afferent neuron envelopes the hair cell, forming an enlarged synaptic terminal called a calyx. The vestibular hair cell-calyx synapse supports a mysterious form of electrical transmission that does not involve gap junctions, termed nonquantal transmission (NQT). The NQT mechanism is thought to involve the flow of ions from the presynaptic hair cell to the postsynaptic calyx through low-voltage-activated channels driven by changes in cleft [K+] as K+ exits the hair cell. However, this hypothesis has not been tested with a quantitative model and the possible role of an electrical potential in the cleft has remained speculative. Here, we present a computational model that captures experimental observations of NQT and identifies features that support the existence of an electrical potential (ϕ) in the synaptic cleft. We show that changes in cleft ϕ reduce transmission latency and illustrate the relative contributions of both cleft [K+] and ϕ to the gain and phase of NQT. We further demonstrate that the magnitude and speed of NQT depend on calyx morphology and that increasing calyx height reduces action potential latency in the calyx afferent. These predictions are consistent with the idea that the calyx evolved to enhance NQT and speed up vestibular signals that drive neural circuits controlling gaze, balance, and orientation.
Collapse
Affiliation(s)
- Aravind Chenrayan Govindaraju
- aApplied Physics Graduate Program, Smalley-Curl Institute, Rice University, Houston, TX77005
- bDepartment of Bioengineering, Rice University, Houston, TX77005
| | - Imran H. Quraishi
- cDepartment of Neurology, Yale University School of Medicine, New Haven, CT06510
| | - Anna Lysakowski
- dDepartment of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL60612
| | - Ruth Anne Eatock
- eDepartment of Neurobiology, University of Chicago, Chicago, IL60637
| | - Robert M. Raphael
- bDepartment of Bioengineering, Rice University, Houston, TX77005
- 1To whom correspondence may be addressed.
| |
Collapse
|
7
|
Contini D, Holstein GR, Art JJ. Simultaneous Dual Recordings From Vestibular Hair Cells and Their Calyx Afferents Demonstrate Multiple Modes of Transmission at These Specialized Endings. Front Neurol 2022; 13:891536. [PMID: 35899268 PMCID: PMC9310783 DOI: 10.3389/fneur.2022.891536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/02/2022] [Indexed: 11/18/2022] Open
Abstract
In the vestibular periphery, transmission via conventional synaptic boutons is supplemented by post-synaptic calyceal endings surrounding Type I hair cells. This review focusses on the multiple modes of communication between these receptors and their enveloping calyces as revealed by simultaneous dual-electrode recordings. Classic orthodromic transmission is accompanied by two forms of bidirectional communication enabled by the extensive cleft between the Type I hair cell and its calyx. The slowest cellular communication low-pass filters the transduction current with a time constant of 10–100 ms: potassium ions accumulate in the synaptic cleft, depolarizing both the hair cell and afferent to potentials greater than necessary for rapid vesicle fusion in the receptor and potentially triggering action potentials in the afferent. On the millisecond timescale, conventional glutamatergic quantal transmission occurs when hair cells are depolarized to potentials sufficient for calcium influx and vesicle fusion. Depolarization also permits a third form of transmission that occurs over tens of microseconds, resulting from the large voltage- and ion-sensitive cleft-facing conductances in both the hair cell and the calyx that are open at their resting potentials. Current flowing out of either the hair cell or the afferent divides into the fraction flowing across the cleft into its cellular partner, and the remainder flowing out of the cleft and into the surrounding fluid compartment. These findings suggest multiple biophysical bases for the extensive repertoire of response dynamics seen in the population of primary vestibular afferent fibers. The results further suggest that evolutionary pressures drive selection for the calyx afferent.
Collapse
Affiliation(s)
- Donatella Contini
- Department of Anatomy & Cell Biology, University of Illinois College of Medicine, Chicago, IL, United States
| | - Gay R. Holstein
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jonathan J. Art
- Department of Anatomy & Cell Biology, University of Illinois College of Medicine, Chicago, IL, United States
- *Correspondence: Jonathan J. Art
| |
Collapse
|
8
|
Mukhopadhyay M, Pangrsic T. Synaptic transmission at the vestibular hair cells of amniotes. Mol Cell Neurosci 2022; 121:103749. [PMID: 35667549 DOI: 10.1016/j.mcn.2022.103749] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 05/09/2022] [Accepted: 06/01/2022] [Indexed: 11/19/2022] Open
Abstract
A harmonized interplay between the central nervous system and the five peripheral end organs is how the vestibular system helps organisms feel a sense of balance and motion in three-dimensional space. The receptor cells of this system, much like their cochlear equivalents, are the specialized hair cells. However, research over the years has shown that the vestibular endorgans and hair cells evolved very differently from their cochlear counterparts. The structurally unique calyceal synapse, which appeared much later in the evolutionary time scale, and continues to intrigue researchers, is now known to support several forms of synaptic neurotransmission. The conventional quantal transmission is believed to employ the ribbon structures, which carry several tethered vesicles filled with neurotransmitters. However, the field of vestibular hair cell synaptic molecular anatomy is still at a nascent stage and needs further work. In this review, we will touch upon the basic structure and function of the peripheral vestibular system, with the focus on the various modes of neurotransmission at the type I vestibular hair cells. We will also shed light on the current knowledge about the molecular anatomy of the vestibular hair cell synapses and vestibular synaptopathy.
Collapse
Affiliation(s)
- Mohona Mukhopadhyay
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, and Institute for Auditory Neuroscience, 37075 Göttingen, Germany
| | - Tina Pangrsic
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, and Institute for Auditory Neuroscience, 37075 Göttingen, Germany; Auditory Neuroscience Group, Max Planck Institute for Multidisciplinary Sciences, 37075 Göttingen, Germany; Collaborative Research Center 889, University of Göttingen, Göttingen, Germany; Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, 37075 Göttingen, Germany.
| |
Collapse
|
9
|
Lee C, Sinha AK, Henry K, Walbaum AW, Crooks PA, Holt JC. Characterizing the Access of Cholinergic Antagonists to Efferent Synapses in the Inner Ear. Front Neurosci 2022; 15:754585. [PMID: 34970112 PMCID: PMC8712681 DOI: 10.3389/fnins.2021.754585] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/15/2021] [Indexed: 11/13/2022] Open
Abstract
Stimulation of cholinergic efferent neurons innervating the inner ear has profound, well-characterized effects on vestibular and auditory physiology, after activating distinct ACh receptors (AChRs) on afferents and hair cells in peripheral endorgans. Efferent-mediated fast and slow excitation of vestibular afferents are mediated by α4β2*-containing nicotinic AChRs (nAChRs) and muscarinic AChRs (mAChRs), respectively. On the auditory side, efferent-mediated suppression of distortion product otoacoustic emissions (DPOAEs) is mediated by α9α10nAChRs. Previous characterization of these synaptic mechanisms utilized cholinergic drugs, that when systemically administered, also reach the CNS, which may limit their utility in probing efferent function without also considering central effects. Use of peripherally-acting cholinergic drugs with local application strategies may be useful, but this approach has remained relatively unexplored. Using multiple administration routes, we performed a combination of vestibular afferent and DPOAE recordings during efferent stimulation in mouse and turtle to determine whether charged mAChR or α9α10nAChR antagonists, with little CNS entry, can still engage efferent synaptic targets in the inner ear. The charged mAChR antagonists glycopyrrolate and methscopolamine blocked efferent-mediated slow excitation of mouse vestibular afferents following intraperitoneal, middle ear, or direct perilymphatic administration. Both mAChR antagonists were effective when delivered to the middle ear, contralateral to the side of afferent recordings, suggesting they gain vascular access after first entering the perilymphatic compartment. In contrast, charged α9α10nAChR antagonists blocked efferent-mediated suppression of DPOAEs only upon direct perilymphatic application, but failed to reach efferent synapses when systemically administered. These data show that efferent mechanisms are viable targets for further characterizing drug access in the inner ear.
Collapse
Affiliation(s)
- Choongheon Lee
- Department of Otolaryngology, University of Rochester, Rochester, NY, United States
| | - Anjali K Sinha
- Department of Neuroscience, University of Rochester, Rochester, NY, United States
| | - Kenneth Henry
- Department of Otolaryngology, University of Rochester, Rochester, NY, United States.,Department of Neuroscience, University of Rochester, Rochester, NY, United States
| | - Anqi W Walbaum
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Peter A Crooks
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Joseph C Holt
- Department of Otolaryngology, University of Rochester, Rochester, NY, United States.,Department of Neuroscience, University of Rochester, Rochester, NY, United States.,Department of Pharmacology & Physiology, University of Rochester, Rochester, NY, United States
| |
Collapse
|
10
|
Meredith FL, Rennie KJ. Dopaminergic Inhibition of Na + Currents in Vestibular Inner Ear Afferents. Front Neurosci 2021; 15:710321. [PMID: 34580582 PMCID: PMC8463658 DOI: 10.3389/fnins.2021.710321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 08/10/2021] [Indexed: 11/13/2022] Open
Abstract
Inner ear hair cells form synapses with afferent terminals and afferent neurons carry signals as action potentials to the central nervous system. Efferent neurons have their origins in the brainstem and some make synaptic contact with afferent dendrites beneath hair cells. Several neurotransmitters have been identified that may be released from efferent terminals to modulate afferent activity. Dopamine is a candidate efferent neurotransmitter in both the vestibular and auditory systems. Within the cochlea, activation of dopamine receptors may reduce excitotoxicity at the inner hair cell synapse via a direct effect of dopamine on afferent terminals. Here we investigated the effect of dopamine on sodium currents in acutely dissociated vestibular afferent calyces to determine if dopaminergic signaling could also modulate vestibular responses. Calyx terminals were isolated along with their accompanying type I hair cells from the cristae of gerbils (P15-33) and whole cell patch clamp recordings performed. Large transient sodium currents were present in all isolated calyces; compared to data from crista slices, resurgent Na+ currents were rare. Perfusion of dopamine (100 μM) in the extracellular solution significantly reduced peak transient Na+ currents by approximately 20% of control. A decrease in Na+ current amplitude was also seen with extracellular application of the D2 dopamine receptor agonist quinpirole, whereas the D2 receptor antagonist eticlopride largely abolished the response to dopamine. Inclusion of the phosphatase inhibitor okadaic acid in the patch electrode solution occluded the response to dopamine. The reduction in calyx sodium current in response to dopamine suggests efferent signaling through D2 dopaminergic receptors may occur via common mechanisms to decrease excitability in inner ear afferents.
Collapse
Affiliation(s)
- Frances L Meredith
- Department of Otolaryngology - Head & Neck Surgery, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Katherine J Rennie
- Department of Otolaryngology - Head & Neck Surgery, School of Medicine, University of Colorado, Aurora, CO, United States.,Department of Physiology & Biophysics, School of Medicine, University of Colorado, Aurora, CO, United States
| |
Collapse
|
11
|
The mammalian efferent vestibular system utilizes cholinergic mechanisms to excite primary vestibular afferents. Sci Rep 2021; 11:1231. [PMID: 33441862 PMCID: PMC7806594 DOI: 10.1038/s41598-020-80367-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/18/2020] [Indexed: 02/06/2023] Open
Abstract
Electrical stimulation of the mammalian efferent vestibular system (EVS) predominantly excites primary vestibular afferents along two distinct time scales. Although roles for acetylcholine (ACh) have been demonstrated in other vertebrates, synaptic mechanisms underlying mammalian EVS actions are not well-characterized. To determine if activation of ACh receptors account for efferent-mediated afferent excitation in mammals, we recorded afferent activity from the superior vestibular nerve of anesthetized C57BL/6 mice while stimulating EVS neurons in the brainstem, before and after administration of cholinergic antagonists. Using a normalized coefficient of variation (CV*), we broadly classified vestibular afferents as regularly- (CV* < 0.1) or irregularly-discharging (CV* > 0.1) and characterized their responses to midline or ipsilateral EVS stimulation. Afferent responses to efferent stimulation were predominantly excitatory, grew in amplitude with increasing CV*, and consisted of fast and slow components that could be identified by differences in rise time and post-stimulus duration. Both efferent-mediated excitatory components were larger in irregular afferents with ipsilateral EVS stimulation. Our pharmacological data show, for the first time in mammals, that muscarinic AChR antagonists block efferent-mediated slow excitation whereas the nicotinic AChR antagonist DHβE selectively blocks efferent-mediated fast excitation, while leaving the efferent-mediated slow component intact. These data confirm that mammalian EVS actions are predominantly cholinergic.
Collapse
|
12
|
Ramakrishna Y, Manca M, Glowatzki E, Sadeghi SG. Cholinergic Modulation of Membrane Properties of Calyx Terminals in the Vestibular Periphery. Neuroscience 2020; 452:98-110. [PMID: 33197502 DOI: 10.1016/j.neuroscience.2020.10.035] [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: 04/21/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 01/10/2023]
Abstract
Vestibular nerve afferents are divided into regular and irregular groups based on the variability of interspike intervals in their resting discharge. Most afferents receive inputs from bouton terminals that contact type II hair cells as well as from calyx terminals that cover the basolateral walls of type I hair cells. Calyces have an abundance of different subtypes of KCNQ (Kv7) potassium channels and muscarinic acetylcholine receptors (mAChRs) and receive cholinergic efferent inputs from neurons in the brainstem. We investigated whether mAChRs affected membrane properties and firing patterns of calyx terminals through modulation of KCNQ channel activity. Patch clamp recordings were performed from calyx terminals in central regions of the cristae of the horizontal and anterior canals in 13-26 day old Sprague-Dawley rats. KCNQ mediated currents were observed as voltage sensitive currents with slow kinetics (activation and deactivation), resulting in spike frequency adaptation so that calyces at best fired a single action potential at the beginning of a depolarizing step. Activation of mAChRs by application of oxotremorine methiodide or inhibition of KCNQ channels by linopirdine dihydrochloride decreased voltage activated currents by ∼30%, decreased first spike latencies by ∼40%, resulted in action potential generation in response to smaller current injections and at lower (i.e., more hyperpolarized) membrane potentials, and increased the number of spikes fired during depolarizing steps. Interestingly, some of the calyces showed spontaneous discharge in the presence of these drugs. Together, these findings suggest that cholinergic efferents can modulate the response properties and encoding of head movements by afferents.
Collapse
Affiliation(s)
- Yugandhar Ramakrishna
- Center for Hearing and Deafness, Department of Communicative Disorders and Sciences, State University of New York at Buffalo, Buffalo, NY, United States; Department of Communication Disorders and Sciences, California State University, Northridge, CA, United States
| | - Marco Manca
- Department of Otolaryngology - Head and Neck Surgery, The Center for Hearing and Balance, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Elisabeth Glowatzki
- Department of Otolaryngology - Head and Neck Surgery, The Center for Hearing and Balance, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Soroush G Sadeghi
- Center for Hearing and Deafness, Department of Communicative Disorders and Sciences, State University of New York at Buffalo, Buffalo, NY, United States; Neuroscience Program, State University of New York at Buffalo, Buffalo, NY, United States.
| |
Collapse
|
13
|
Ramakrishna Y, Sadeghi SG. Activation of GABA B receptors results in excitatory modulation of calyx terminals in rat semicircular canal cristae. J Neurophysiol 2020; 124:962-972. [PMID: 32816581 PMCID: PMC7509296 DOI: 10.1152/jn.00243.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 12/15/2022] Open
Abstract
Previous studies have found GABA in vestibular end organs. However, existence of GABA receptors or possible GABAergic effects on vestibular nerve afferents has not been investigated. The current study was conducted to determine whether activation of GABAB receptors affects calyx afferent terminals in the central region of the cristae of semicircular canals. We used patch-clamp recording in postnatal day 13-18 (P13-P18) Sprague-Dawley rats of either sex. Application of GABAB receptor agonist baclofen inhibited voltage-sensitive potassium currents. This effect was blocked by selective GABAB receptor antagonist CGP 35348. Application of antagonists of small (SK)- and large-conductance potassium (BK) channels almost completely blocked the effects of baclofen. The remaining baclofen effect was blocked by cadmium chloride, suggesting that it could be due to inhibition of voltage-gated calcium channels. Furthermore, baclofen had no effect in the absence of calcium in the extracellular fluid. Inhibition of potassium currents by GABAB activation resulted in an excitatory effect on calyx terminal action potential firing. While in the control condition calyces could only fire a single action potential during step depolarizations, in the presence of baclofen they fired continuously during steps and a few even showed repetitive discharge. We also found a decrease in threshold for action potential generation and a decrease in first-spike latency during step depolarization. These results provide the first evidence for the presence of GABAB receptors on calyx terminals, showing that their activation results in an excitatory effect and that GABA inputs could be used to modulate calyx response properties.NEW & NOTEWORTHY Using in vitro whole cell patch-clamp recordings from calyx terminals in the vestibular end organs, we show that activation of GABAB receptors result in an excitatory effect, with decreased spike-frequency adaptation and shortened first-spike latencies. Our results suggest that these effects are mediated through inhibition of calcium-sensitive potassium channels.
Collapse
Affiliation(s)
- Yugandhar Ramakrishna
- Center for Hearing and Deafness, Department of Communicative Disorders and Sciences, State University of New York at Buffalo, Buffalo, New York
- Department of Communication Disorders and Sciences, California State University, Northridge, Northridge, California
| | - Soroush G Sadeghi
- Center for Hearing and Deafness, Department of Communicative Disorders and Sciences, State University of New York at Buffalo, Buffalo, New York
- Neuroscience Program, State University of New York at Buffalo, Buffalo, New York
| |
Collapse
|
14
|
Holman HA, Wan Y, Rabbitt RD. Developmental GAD2 Expression Reveals Progenitor-like Cells with Calcium Waves in Mammalian Crista Ampullaris. iScience 2020; 23:101407. [PMID: 32771977 PMCID: PMC7415930 DOI: 10.1016/j.isci.2020.101407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/29/2020] [Accepted: 07/21/2020] [Indexed: 01/26/2023] Open
Abstract
Sense of motion, spatial orientation, and balance in vertebrates relies on sensory hair cells in the inner ear vestibular system. Vestibular supporting cells can regenerate hair cells that are lost from aging, ototoxicity, and trauma, although not all factors or specific cell types are known. Here we report a population of GAD2-positive cells in the mouse crista ampullaris and trace GAD2 progenitor-like cells that express pluripotent transcription factors SOX2, PROX1, and CTBP2. GAD2 progenitor-like cells organize into rosettes around a central branched structure in the eminentia cruciatum (EC) herein named the EC plexus. GCaMP5G calcium indicator shows spontaneous and acetylcholine-evoked whole-cell calcium waves in neonatal and adult mice. We present a hypothetical model that outlines the lineage and potential regenerative capacity of GAD2 cells in the mammalian vestibular neuroepithelium.
Collapse
Affiliation(s)
- Holly A Holman
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| | - Yong Wan
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Richard D Rabbitt
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA; Graduate Program in Neuroscience, University of Utah, Salt Lake City, UT 84112, USA; Department of Otolaryngology-Head & Neck Surgery, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
15
|
Yu Z, McIntosh JM, Sadeghi SG, Glowatzki E. Efferent synaptic transmission at the vestibular type II hair cell synapse. J Neurophysiol 2020; 124:360-374. [PMID: 32609559 DOI: 10.1152/jn.00143.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In the vestibular peripheral organs, type I and type II hair cells (HCs) transmit incoming signals via glutamatergic quantal transmission onto afferent nerve fibers. Additionally, type I HCs transmit via "non-quantal" transmission to calyx afferent fibers, by accumulation of glutamate and potassium in the synaptic cleft. Vestibular efferent inputs originating in the brainstem contact type II HCs and vestibular afferents. Here, synaptic inputs to type II HCs were characterized by using electrical and optogenetic stimulation of efferent fibers combined with in vitro whole cell patch-clamp recording from type II HCs in the rodent vestibular crista. Properties of efferent synaptic currents in type II HCs were similar to those found in cochlear HCs and mediated by activation of α9-containing nicotinic acetylcholine receptors (nAChRs) and small-conductance calcium-activated potassium (SK) channels. While efferents showed a low probability of release at low frequencies of stimulation, repetitive stimulation resulted in facilitation and increased probability of release. Notably, the membrane potential of type II HCs during optogenetic stimulation of efferents showed a strong hyperpolarization in response to single pulses and was further enhanced by repetitive stimulation. Such efferent-mediated inhibition of type II HCs can provide a mechanism to adjust the contribution of signals from type I and type II HCs to vestibular nerve fibers, with a shift of the response to be more like that of calyx-only afferents with faster non-quantal responses.NEW & NOTEWORTHY Type II vestibular hair cells (HCs) receive inputs from efferent neurons in the brain stem. We used in vitro optogenetic and electrical stimulation of vestibular efferent fibers to study their synaptic inputs to type II HCs. Stimulation of efferents inhibited type II HCs, similar to efferent effects on cochlear HCs. We propose that efferent inputs adjust the contribution of signals from type I and II HCs to vestibular nerve fibers.
Collapse
Affiliation(s)
- Zhou Yu
- Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Otolaryngology-Head and Neck Surgery, The Center for Hearing and Balance, and The Center for Sensory Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Soroush G Sadeghi
- Department of Communicative Disorders and Sciences, and Center for Hearing and Deafness, State University of New York at Buffalo, Buffalo, New York.,Neuroscience Program, State University of New York at Buffalo, Buffalo, New York
| | - Elisabeth Glowatzki
- Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Otolaryngology-Head and Neck Surgery, The Center for Hearing and Balance, and The Center for Sensory Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
16
|
Longridge NS, Lim A, Mallinson AI, Renshaw J. Vestibular suppression of normal bodily sounds. Acta Otolaryngol 2020; 140:401-405. [PMID: 32068478 DOI: 10.1080/00016489.2020.1723807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Background: Vestibular-evoked myogenic potentials (VEMPs) are present in almost everyone. The proximity of the cochlear and vestibular end organs strongly indicates an overlap of the stimuli to which they respond.Aims/objectives: To determine the loudness of shouting compared to the external auditory canal (EAC) and explore how hyperstimulation of the cochlea and vestibular structures might be prevented.Materials and methods: We compared the loudness and frequency response of sound during shouting, biting and chewing, as well as the measurement of the loudness and frequency of sound in the external auditory canal (EAC) when a calibrated minishaker is applied to the forehead and to the upper incisor teeth.Results: We showed that vibratory sounds produced by vocalizations and oral activities were attenuated when they reached the ear, so that these sounds would not act as vestibular stimulants.Conclusions and significance: Chewing is known to cause a stapedius reflex which suppresses internal sounds to optimize audition of external sounds, while at the same time suppressing vestibular stimulation, which serves to optimize the sensitivity of the vestibular system, in order that it may respond precisely to a threat.
Collapse
Affiliation(s)
- Neil S. Longridge
- Division of Otolaryngology, Department of Surgery, The University of British Columbia, Vancouver, Canada
| | - Anielle Lim
- Department of Audiology, Providence Health Care, Vancouver, Canada
| | - Arthur Ian Mallinson
- Division of Otolaryngology, Department of Surgery, The University of British Columbia, Vancouver, Canada
- Neuro-otology Unit, Vancouver General Hospital, Vancouver, Canada
| | - Jim Renshaw
- Neuro-otology Unit, Vancouver General Hospital, Vancouver, Canada
| |
Collapse
|
17
|
Longridge NS. Vestibular evoked myogenic potentials: what are they for? An opinion; a hypothesis. Acta Otolaryngol 2020; 140:255-257. [PMID: 32149556 DOI: 10.1080/00016489.2019.1704545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 11/02/2019] [Indexed: 10/24/2022]
Affiliation(s)
- Neil S Longridge
- Division of Otolaryngology, Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| |
Collapse
|
18
|
Spaiardi P, Tavazzani E, Manca M, Russo G, Prigioni I, Biella G, Giunta R, Johnson SL, Marcotti W, Masetto S. K + Accumulation and Clearance in the Calyx Synaptic Cleft of Type I Mouse Vestibular Hair Cells. Neuroscience 2020; 426:69-86. [PMID: 31846752 PMCID: PMC6985899 DOI: 10.1016/j.neuroscience.2019.11.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 11/07/2019] [Accepted: 11/16/2019] [Indexed: 11/29/2022]
Abstract
Vestibular organs of Amniotes contain two types of sensory cells, named Type I and Type II hair cells. While Type II hair cells are contacted by several small bouton nerve terminals, Type I hair cells receive a giant terminal, called a calyx, which encloses their basolateral membrane almost completely. Both hair cell types release glutamate, which depolarizes the afferent terminal by binding to AMPA post-synaptic receptors. However, there is evidence that non-vesicular signal transmission also occurs at the Type I hair cell-calyx synapse, possibly involving direct depolarization of the calyx by K+ exiting the hair cell. To better investigate this aspect, we performed whole-cell patch-clamp recordings from mouse Type I hair cells or their associated calyx. We found that [K+] in the calyceal synaptic cleft is elevated at rest relative to the interstitial (extracellular) solution and can increase or decrease during hair cell depolarization or repolarization, respectively. The change in [K+] was primarily driven by GK,L, the low-voltage-activated, non-inactivating K+ conductance specifically expressed by Type I hair cells. Simple diffusion of K+ between the cleft and the extracellular compartment appeared substantially restricted by the calyx inner membrane, with the ion channels and active transporters playing a crucial role in regulating intercellular [K+]. Calyx recordings were consistent with K+ leaving the synaptic cleft through postsynaptic voltage-gated K+ channels involving KV1 and KV7 subunits. The above scenario is consistent with direct depolarization and hyperpolarization of the calyx membrane potential by intercellular K+.
Collapse
Affiliation(s)
- P Spaiardi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia 27100, Italy
| | - E Tavazzani
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia 27100, Italy
| | - M Manca
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia 27100, Italy
| | - G Russo
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia 27100, Italy
| | - I Prigioni
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia 27100, Italy
| | - G Biella
- Department of Biology and Biotechnology, University of Pavia, Pavia 27100, Italy
| | - R Giunta
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia 27100, Italy
| | - S L Johnson
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| | - W Marcotti
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| | - S Masetto
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia 27100, Italy.
| |
Collapse
|
19
|
Affiliation(s)
- Henrique von Gersdorff
- Vollum Institute, Oregon Health & Science University, Portland, Oregon, 97239, USA.,Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, 97239, USA.,Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon, 97239, USA
| | - Marta M Iversen
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, 84112, USA
| | - Richard D Rabbitt
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, 84112, USA.,Department of Otolaryngology, University of Utah, Salt Lake City, Utah, 84112, USA
| |
Collapse
|
20
|
Poppi LA, Holt JC, Lim R, Brichta AM. A review of efferent cholinergic synaptic transmission in the vestibular periphery and its functional implications. J Neurophysiol 2019; 123:608-629. [PMID: 31800345 DOI: 10.1152/jn.00053.2019] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
It has been over 60 years since peripheral efferent vestibular terminals were first identified in mammals, and yet the function of the efferent vestibular system remains obscure. One reason for the lack of progress may be due to our deficient understanding of the peripheral efferent synapse. Although vestibular efferent terminals were identified as cholinergic less than a decade after their anatomical characterization, the cellular mechanisms that underlie the properties of these synapses have had to be inferred. In this review we examine how recent mammalian studies have begun to reveal both nicotinic and muscarinic effects at these terminals and therefore provide a context for fast and slow responses observed in classic electrophysiological studies of the mammalian efferent vestibular system, nearly 40 years ago. Although incomplete, these new results together with those of recent behavioral studies are helping to unravel the mysterious and perplexing action of the efferent vestibular system. Armed with this information, we may finally appreciate the behavioral framework in which the efferent vestibular system operates.
Collapse
Affiliation(s)
- L A Poppi
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,Preclinical Neurobiology Research Group, The University of Newcastle, Newcastle, NSW, Australia
| | - J C Holt
- Department of Otolaryngology, University of Rochester Medical Center, Rochester, New York
| | - R Lim
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,Preclinical Neurobiology Research Group, The University of Newcastle, Newcastle, NSW, Australia
| | - A M Brichta
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,Preclinical Neurobiology Research Group, The University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
21
|
Efferent Inputs Are Required for Normal Function of Vestibular Nerve Afferents. J Neurosci 2019; 39:6922-6935. [PMID: 31285300 DOI: 10.1523/jneurosci.0237-19.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 06/28/2019] [Accepted: 07/01/2019] [Indexed: 01/09/2023] Open
Abstract
A group of vestibular afferent nerve fibers with irregular-firing resting discharges are thought to play a prominent role in responses to fast head movements and vestibular plasticity. We show that, in C57BL/6 mice (either sex, 4-5 weeks old), normal activity in the efferent vestibular pathway is required for function of these irregular afferents. Thermal inhibition of efferent fibers results in a profound inhibition of irregular afferents' resting discharges, rendering them inadequate for signaling head movements. In this way, efferent inputs adjust the contribution of the peripheral irregular afferent pathway that plays a critical role in peripheral vestibular signaling and plasticity.SIGNIFICANCE STATEMENT Vestibular end organs in the inner ear receive efferent inputs from the brainstem. Previously, electrical stimulation of efferents was linked to an increase in resting discharges of afferents and a decrease in their sensitivities. Here, we show that localized thermal inhibition of unmyelinated efferents results in a significant decrease in the activity of afferent nerve fibers, particularly those with irregular resting discharges implicated in responses to fast head movements and vestibular compensation. Thus, by upregulating and downregulating of afferent firing, particularly irregular afferents, efferents adjust neural activity sensitive to rapid head movements. These findings support the notion that peripheral vestibular end organs are not passive transducers of head movements and their sensory signal transmission is modulated by efferent inputs.
Collapse
|
22
|
Holman HA, Poppi LA, Frerck M, Rabbitt RD. Spontaneous and Acetylcholine Evoked Calcium Transients in the Developing Mouse Utricle. Front Cell Neurosci 2019; 13:186. [PMID: 31133810 PMCID: PMC6514437 DOI: 10.3389/fncel.2019.00186] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 04/12/2019] [Indexed: 11/13/2022] Open
Abstract
Spontaneous calcium transients are present during early postnatal development in the mouse retina and cochlea, and play an important role in maturation of the sensory organs and neural circuits in the central nervous system (CNS). It is not known whether similar calcium transients occur during postnatal development in the vestibular sensory organs. Here we demonstrate spontaneous intracellular calcium transients in sensory hair cells (HCs) and supporting cells (SCs) in the murine utricular macula during the first two postnatal weeks. Calcium transients were monitored using a genetically encoded calcium indicator, GCaMP5G (G5), at 100 ms-frame−1 in excised utricle sensory epithelia, including HCs, SCs, and neurons. The reporter line expressed G5 and tdTomato (tdT) in a Gad2-Cre dependent manner within a subset of utricular HCs, SCs and neurons. Kinetics of the G5 reporter limited temporal resolution to calcium events lasting longer than 200 ms. Spontaneous calcium transients lasting 1-2 s were observed in the expressing population of HCs at birth and slower spontaneous transients lasting 10-30 s appeared in SCs by P3. Beginning at P5, calcium transients could be modulated by application of the efferent neurotransmitter acetylcholine (ACh). In mature mice, calcium transients in the utricular macula occurred spontaneously, had a duration 1-2 s, and could be modulated by the exogenous application of acetylcholine (ACh) or muscarine. Long-lasting calcium transients evoked by ACh in mature mice were blocked by atropine, consistent with previous reports describing the role of muscarinic receptors expressed in calyx bearing afferents in efferent control of vestibular sensation. Large spontaneous and ACh evoked transients were reversibly blocked by the inositol trisphosphate receptor (IP3R) antagonist aminoethoxydiphenyl borate (2-APB). Results demonstrate long-lasting calcium transients are present in the utricular macula during the first postnatal week, and that responses to ACh mature over this same time period.
Collapse
Affiliation(s)
- Holly A Holman
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Lauren A Poppi
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States.,School of Biomedical Science and Pharmacy, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | - Micah Frerck
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States
| | - Richard D Rabbitt
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, United States.,Neuroscience Program, University of Utah, Salt Lake City, UT, United States.,Otolaryngology-Head and Neck Surgery, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
23
|
Rabbitt RD. Semicircular canal biomechanics in health and disease. J Neurophysiol 2019; 121:732-755. [PMID: 30565972 PMCID: PMC6520623 DOI: 10.1152/jn.00708.2018] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 12/11/2018] [Accepted: 12/11/2018] [Indexed: 12/12/2022] Open
Abstract
The semicircular canals are responsible for sensing angular head motion in three-dimensional space and for providing neural inputs to the central nervous system (CNS) essential for agile mobility, stable vision, and autonomic control of the cardiovascular and other gravity-sensitive systems. Sensation relies on fluid mechanics within the labyrinth to selectively convert angular head acceleration into sensory hair bundle displacements in each of three inner ear sensory organs. Canal afferent neurons encode the direction and time course of head movements over a broad range of movement frequencies and amplitudes. Disorders altering canal mechanics result in pathological inputs to the CNS, often leading to debilitating symptoms. Vestibular disorders and conditions with mechanical substrates include benign paroxysmal positional nystagmus, direction-changing positional nystagmus, alcohol positional nystagmus, caloric nystagmus, Tullio phenomena, and others. Here, the mechanics of angular motion transduction and how it contributes to neural encoding by the semicircular canals is reviewed in both health and disease.
Collapse
Affiliation(s)
- R. D. Rabbitt
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah
- Otolaryngology-Head Neck Surgery, University of Utah, Salt Lake City, Utah
- Neuroscience Program, University of Utah, Salt Lake City, Utah
| |
Collapse
|
24
|
Eatock RA. Specializations for Fast Signaling in the Amniote Vestibular Inner Ear. Integr Comp Biol 2019; 58:341-350. [PMID: 29920589 DOI: 10.1093/icb/icy069] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
During rapid locomotion, the vestibular inner ear provides head-motion signals that stabilize posture, gaze, and heading. Afferent nerve fibers from central and peripheral zones of vestibular sensory epithelia use temporal and rate encoding, respectively, to emphasize different aspects of head motion: central afferents adapt faster to sustained head position and favor higher stimulus frequencies, reflecting specializations at each stage from motion of the accessory structure to spike propagation to the brain. One specialization in amniotes is an unusual nonquantal synaptic mechanism by which type I hair cells transmit to large calyceal terminals of afferent neurons. The reduced synaptic delay of this mechanism may have evolved to serve reliable and fast input to reflex pathways that ensure stable locomotion on land.
Collapse
Affiliation(s)
- Ruth Anne Eatock
- Department of Neurobiology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
25
|
Enhanced Activation of HCN Channels Reduces Excitability and Spike-Timing Regularity in Maturing Vestibular Afferent Neurons. J Neurosci 2019; 39:2860-2876. [PMID: 30696730 DOI: 10.1523/jneurosci.1811-18.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 01/14/2019] [Accepted: 01/19/2019] [Indexed: 11/21/2022] Open
Abstract
Vestibular ganglion neurons (VGNs) transmit information along parallel neuronal pathways whose signature distinction is variability in spike-timing; some fire at regular intervals while others fire at irregular intervals. The mechanisms driving timing differences are not fully understood but two opposing (but not mutually exclusive) hypotheses have emerged. In the first, regular-spiking is inversely correlated to the density of low-voltage-gated potassium currents (I KL). In the second, regular spiking is directly correlated to the density of hyperpolarization-activated cyclic nucleotide-sensitive currents (I H). Supporting the idea that variations in ion channel composition shape spike-timing, VGNs from the first postnatal week respond to synaptic-noise-like current injections with irregular-firing patterns if they have I KL and with more regular firing patterns if they do not. However, in vitro firing patterns are not as regular as those in vivo Here we considered whether highly-regular spiking requires I H currents and whether this dependence emerges later in development after channel expression matures. We recorded from rat VGN somata of either sex aged postnatal day (P)9-P21. Counter to expectation, in vitro firing patterns were less diverse, more transient-spiking, and more irregular at older ages than at younger ages. Resting potentials hyperpolarized and resting conductance increased, consistent with developmental upregulation of I KL Activation of I H (by increasing intracellular cAMP) increased spike rates but not spike-timing regularity. In a model, we found that activating I H counter-intuitively suppressed regularity by recruiting I KL Developmental upregulation in I KL appears to overwhelm I H These results counter previous hypotheses about how I H shapes vestibular afferent responses.SIGNIFICANCE STATEMENT Vestibular sensory information is conveyed on parallel neuronal pathways with irregularly-firing neurons encoding information using a temporal code and regularly-firing neurons using a rate code. This is a striking example of spike-timing statistics influencing information coding. Previous studies from immature vestibular ganglion neurons (VGNs) identified hyperpolarization-activated mixed cationic currents (I H) as driving highly-regular spiking and proposed that this influence grows with the current during maturation. We found that I H becomes less influential, likely because maturing VGNs also acquire low-voltage-gated potassium currents (I KL), whose inhibitory influence opposes I H Because efferent activity can partly close I KL, VGN firing patterns may become more receptive to extrinsic control. Spike-timing regularity likely relies on dynamic ion channel properties and complementary specializations in synaptic connectivity.
Collapse
|
26
|
Jones SM, Vijayakumar S, Dow SA, Holt JC, Jordan PM, Luebke AE. Loss of α-Calcitonin Gene-Related Peptide (αCGRP) Reduces Otolith Activation Timing Dynamics and Impairs Balance. Front Mol Neurosci 2018; 11:289. [PMID: 30197585 PMCID: PMC6117397 DOI: 10.3389/fnmol.2018.00289] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 07/31/2018] [Indexed: 12/03/2022] Open
Abstract
Calcitonin gene-related peptide (CGRP) is a neuroactive peptide that is thought to play a role at efferent synapses in hair cell organs including the cochlea, lateral line, and semicircular canal. The deletion of CGRP in transgenic mice is associated with a significant reduction in suprathreshold cochlear nerve activity and vestibulo–ocular reflex (VOR) gain efficacy when compared to littermate controls. Here we asked whether the loss of CGRP also influences otolithic end organ function and contributes to balance impairments. Immunostaining for CGRP was absent in the otolithic end organs of αCGRP null (-/-) mice while choline acetyltransferase (ChAT) immunolabeling appeared unchanged suggesting the overall gross development of efferent innervation in otolithic organs was unaltered. Otolithic function was assessed by quantifying the thresholds, suprathreshold amplitudes, and latencies of vestibular sensory-evoked potentials (VsEPs) while general balance function was assessed using a modified rotarod assay. The loss of αCGRP in null (-/-) mice was associated with: (1) shorter VsEP latencies without a concomitant change in amplitude or thresholds, and (2) deficits in the rotarod balance assay. Our findings show that CGRP loss results in faster otolith afferent activation timing, suggesting that the CGRP component of the efferent vestibular system (EVS) also plays a role in otolithic organ dynamics, which when coupled with reduced VOR gain efficacy, impairs balance.
Collapse
Affiliation(s)
- Sherri M Jones
- Department of Special Education and Communication Disorders, University of Nebraska, Lincoln, NE, United States
| | - Sarath Vijayakumar
- Department of Special Education and Communication Disorders, University of Nebraska, Lincoln, NE, United States
| | - Samantha A Dow
- Department of Neuroscience and Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, United States
| | - Joseph C Holt
- Department of Neuroscience and Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, United States.,Department of Otolaryngology, University of Rochester Medical Center, Rochester, NY, United States
| | - Paivi M Jordan
- Department of Otolaryngology, University of Rochester Medical Center, Rochester, NY, United States
| | - Anne E Luebke
- Department of Neuroscience and Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, United States.,Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
27
|
Abstract
Sensory hair cells are specialized secondary sensory cells that mediate our senses of hearing, balance, linear acceleration, and angular acceleration (head rotation). In addition, hair cells in fish and amphibians mediate sensitivity to water movement through the lateral line system, and closely related electroreceptive cells mediate sensitivity to low-voltage electric fields in the aquatic environment of many fish species and several species of amphibian. Sensory hair cells share many structural and functional features across all vertebrate groups, while at the same time they are specialized for employment in a wide variety of sensory tasks. The complexity of hair cell structure is large, and the diversity of hair cell applications in sensory systems exceeds that seen for most, if not all, sensory cell types. The intent of this review is to summarize the more significant structural features and some of the more interesting and important physiological mechanisms that have been elucidated thus far. Outside vertebrates, hair cells are only known to exist in the coronal organ of tunicates. Electrical resonance, electromotility, and their exquisite mechanical sensitivity all contribute to the attractiveness of hair cells as a research subject.
Collapse
|
28
|
Pastras CJ, Curthoys IS, Sokolic L, Brown DJ. Suppression of the vestibular short-latency evoked potential by electrical stimulation of the central vestibular system. Hear Res 2018; 361:23-35. [DOI: 10.1016/j.heares.2018.01.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 01/15/2018] [Accepted: 01/22/2018] [Indexed: 01/12/2023]
|
29
|
Guo CK. Existence of nicotinic receptors in a subset of type I vestibular hair cells of guinea pigs. Brain Res 2018; 1681:85-90. [PMID: 29294348 DOI: 10.1016/j.brainres.2017.12.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 11/19/2017] [Accepted: 12/27/2017] [Indexed: 10/18/2022]
Abstract
In mammals, vestibular hair cells (VHCs) are classified as type I and II according to morphological criteria. Acetylcholine (ACh) is identified as the primary efferent neurotransmitter. To date, cholinergic activities have been reported in mammalian type II VHCs, but similar activities in type I VHCs have not been pursued presumably because the body of type I VHCs were suggested to be totally surrounded by afferent nerve calyces. A few reports showed that part of type I VHCs were incompletely surrounded by calyces and received contact from the efferent nerve endings in the mammals studied. The possibility of the expression of cholinergic receptors, their subunit composition, and their function in mammals' type I VHCs are still unclear. In this study, nicotinic responses were investigated by the whole-cell patch clamp technique in isolated type I VHCs of guinea pigs. Of the cells, 7.3% were sensitive to cholinergic agonists and showed an excitatory current at -40 mV which was not sensitive to nifedipine, iberiotoxin (IBTX), and apamin. The main carriers of this current were Na+ and K+. The rank order of activation potency was nicotine > 1,1-dimethyl-4-phenyl-piperazinium (DMPP) > ACh. These nicotinic ACh receptors (nAChRs) were not blocked by strychnine and α-bungarotoxin (α-BTX), but sensitive to d-tubocurarine (dTC) and mecamylamine (Mec). The findings provide physiological evidence that some subtypes of nAChRs may be located in a subset of type I VHCs, which were different from α9α10 nAChRs.
Collapse
Affiliation(s)
- Chang-Kai Guo
- Shoujia ENT Hospital, 180 Jiefang Avenue, Qiaokou District, Wuhan 430033, China.
| |
Collapse
|
30
|
Parks XX, Contini D, Jordan PM, Holt JC. Confirming a Role for α9nAChRs and SK Potassium Channels in Type II Hair Cells of the Turtle Posterior Crista. Front Cell Neurosci 2017; 11:356. [PMID: 29200999 PMCID: PMC5696599 DOI: 10.3389/fncel.2017.00356] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 10/27/2017] [Indexed: 11/17/2022] Open
Abstract
In turtle posterior cristae, cholinergic vestibular efferent neurons (VENs) synapse on type II hair cells, bouton afferents innervating type II hair cells, and afferent calyces innervating type I hair cells. Electrical stimulation of VENs releases acetylcholine (ACh) at these synapses to exert diverse effects on afferent background discharge including rapid inhibition of bouton afferents and excitation of calyx-bearing afferents. Efferent-mediated inhibition is most pronounced in bouton afferents innervating type II hair cells near the torus, but becomes progressively smaller and briefer when moving longitudinally through the crista toward afferents innervating the planum. Sharp-electrode recordings have inferred that efferent-mediated inhibition of bouton afferents requires the sequential activation of alpha9-containing nicotinic ACh receptors (α9*nAChRs) and small-conductance, calcium-dependent potassium channels (SK) in type II hair cells. Gradations in the strength of efferent-mediated inhibition across the crista likely reflect variations in α9*nAChRs and/or SK activation in type II hair cells from those different regions. However, in turtle cristae, neither inference has been confirmed with direct recordings from type II hair cells. To address these gaps, we performed whole-cell, patch-clamp recordings from type II hair cells within a split-epithelial preparation of the turtle posterior crista. Here, we can easily visualize and record hair cells while maintaining their native location within the neuroepithelium. Consistent with α9*nAChR/SK activation, ACh-sensitive currents in type II hair cells were inward at hyperpolarizing potentials but reversed near −90 mV to produce outward currents that typically peaked around −20 mV. ACh-sensitive currents were largest in torus hair cells but absent from hair cells near the planum. In current clamp recordings under zero-current conditions, ACh robustly hyperpolarized type II hair cells. ACh-sensitive responses were reversibly blocked by the α9nAChR antagonists ICS, strychnine, and methyllycaconitine as well as the SK antagonists apamin and UCL1684. Intact efferent terminals in the split-epithelial preparation spontaneously released ACh that also activated α9*nAChRs/SK in type II hair cells. These release events were accelerated with high-potassium external solution and all events were blocked by strychnine, ICS, methyllycaconitine, and apamin. These findings provide direct evidence that activation of α9*nAChR/SK in turtle type II hair cells underlies efferent-mediated inhibition of bouton afferents.
Collapse
Affiliation(s)
- Xiaorong Xu Parks
- Department of Otolaryngology, University of Rochester, Rochester, NY, United States
| | - Donatella Contini
- Department of Otolaryngology, University of Rochester, Rochester, NY, United States
| | - Paivi M Jordan
- Department of Otolaryngology, University of Rochester, Rochester, NY, United States
| | - Joseph C Holt
- Department of Otolaryngology, University of Rochester, Rochester, NY, United States.,Department of Neuroscience, University of Rochester, Rochester, NY, United States.,Department of Pharmacology and Physiology, University of Rochester, Rochester, NY, United States
| |
Collapse
|
31
|
Lee C, Holt JC, Jones TA. Effect of M-current modulation on mammalian vestibular responses to transient head motion. J Neurophysiol 2017; 118:2991-3006. [PMID: 28855291 DOI: 10.1152/jn.00384.2017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 08/25/2017] [Accepted: 08/25/2017] [Indexed: 12/26/2022] Open
Abstract
The precise role and mechanisms underlying efferent modulation of peripheral vestibular afferent function are not well understood in mammals. Clarifying the details of efferent action may lead to new strategies for clinical management of debilitating disturbances in vestibular and balance function. Recent evidence in turtle indicates that efferent modulation of M-currents is likely one mechanism for modifying afferent discharge. M-currents depend in part on KCNQ potassium conductances (Kv7), which can be adjusted through efferent activation of M1, M3, and/or M5 muscarinic acetylcholine receptors (mAChRs). How KCNQ channels and altered M-currents affect vestibular afferent function in vivo is unclear, and whether such a mechanism operates in mammals is unknown. In this study we used the KCNQ antagonist XE991 and the KCNQ activator retigabine in anesthetized mice to evaluate the effects of M-current modulation on peripheral vestibular responses to transient head motion. At low doses of XE991, responses were modestly enhanced, becoming larger in amplitude and shorter in latency. Higher doses of XE991 produced transient response enhancement, followed by steady-state suppression where latencies and thresholds increased and amplitudes decreased. Retigabine produced opposite effects. Auditory function was also impacted, based on results of companion auditory brain stem response testing. We propose that closure of KCNQ channels transforms vestibular afferent behavior by suppressing responses to transient high-frequency stimuli while simultaneously enhancing responses to sustained low-frequency stimulation. Our results clearly demonstrate that KCNQ channels are critical for normal mammalian vestibular function and suggest that efferent action may utilize these mechanisms to modulate the dynamic characteristics and gain of vestibular afferent responses.NEW & NOTEWORTHY The role of calyceal KCNQ channels and associated M-current in normal mammalian vestibular function is unknown. Our results show that calyceal KCNQ channels are critical for normal vestibular function in the intact mammal. The findings provide evidence that efferent modulation of M-currents may act normally to differentially adjust the sensitivity of vestibular neurons to transient and tonic stimulation and that such mechanisms may be targeted to achieve effective clinical management of vestibular disorders.
Collapse
Affiliation(s)
- Choongheon Lee
- Department of Special Education and Communication Disorders, University of Nebraska-Lincoln, Lincoln, Nebraska; and
| | - J Chris Holt
- Department of Otolaryngology, Department of Neuroscience, and Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York
| | - Timothy A Jones
- Department of Special Education and Communication Disorders, University of Nebraska-Lincoln, Lincoln, Nebraska; and
| |
Collapse
|
32
|
Mathews MA, Camp AJ, Murray AJ. Reviewing the Role of the Efferent Vestibular System in Motor and Vestibular Circuits. Front Physiol 2017; 8:552. [PMID: 28824449 PMCID: PMC5539236 DOI: 10.3389/fphys.2017.00552] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 07/17/2017] [Indexed: 12/31/2022] Open
Abstract
Efferent circuits within the nervous system carry nerve impulses from the central nervous system to sensory end organs. Vestibular efferents originate in the brainstem and terminate on hair cells and primary afferent fibers in the semicircular canals and otolith organs within the inner ear. The function of this efferent vestibular system (EVS) in vestibular and motor coordination though, has proven difficult to determine, and remains under debate. We consider current literature that implicate corollary discharge from the spinal cord through the efferent vestibular nucleus (EVN), and hint at a potential role in overall vestibular plasticity and compensation. Hypotheses range from differentiating between passive and active movements at the level of vestibular afferents, to EVS activation under specific behavioral and environmental contexts such as arousal, predation, and locomotion. In this review, we summarize current knowledge of EVS circuitry, its effects on vestibular hair cell and primary afferent activity, and discuss its potential functional roles.
Collapse
Affiliation(s)
- Miranda A Mathews
- Sensory Systems and Integration Laboratory, Bosch Institute, Discipline of Biomedical Science, University of SydneySydney, NSW, Australia
| | - Aaron J Camp
- Sensory Systems and Integration Laboratory, Bosch Institute, Discipline of Biomedical Science, University of SydneySydney, NSW, Australia
| | - Andrew J Murray
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College LondonLondon, United Kingdom
| |
Collapse
|
33
|
Sultemeier DR, Choy KR, Schweizer FE, Hoffman LF. Spaceflight-induced synaptic modifications within hair cells of the mammalian utricle. J Neurophysiol 2017; 117:2163-2178. [PMID: 28228581 DOI: 10.1152/jn.00240.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 02/21/2017] [Accepted: 02/21/2017] [Indexed: 02/08/2023] Open
Abstract
Exposure to the microgravity conditions of spaceflight alleviates the load normally imposed by the Earth's gravitational field on the inner ear utricular epithelia. Previous ultrastructural investigations have shown that spaceflight induces an increase in synapse density within hair cells of the rat utricle. However, the utricle exhibits broad physiological heterogeneity across different epithelial regions, and it is unknown whether capabilities for synaptic plasticity generalize to hair cells across its topography. To achieve systematic and broader sampling of the epithelium than was previously conducted, we used immunohistochemistry and volumetric image analyses to quantify synapse distributions across representative utricular regions in specimens from mice exposed to spaceflight (a 15-day mission of the space shuttle Discovery). These measures were compared with similarly sampled Earth-bound controls. Following paraformaldehyde fixation and microdissection, immunohistochemistry was performed on intact specimens to label presynaptic ribbons (anti-CtBP2) and postsynaptic receptor complexes (anti-Shank1A). Synapses were identified as closely apposed pre- and postsynaptic puncta. Epithelia from horizontal semicircular canal cristae served as "within-specimen" controls, whereas utricles and cristae from Earth-bound cohorts served as experimental controls. We found that synapse densities decreased in the medial extrastriolae of microgravity specimens compared with experimental controls, whereas they were unchanged in the striolae and horizontal cristae from the two conditions. These data demonstrate that structural plasticity was topographically localized to the utricular region that encodes very low frequency and static changes in linear acceleration, and illuminates the remarkable capabilities of utricular hair cells for synaptic plasticity in adapting to novel gravitational environments.NEW & NOTEWORTHY Spaceflight imposes a radically different sensory environment from that in which the inner ear utricle normally operates. We investigated synaptic modifications in utricles from mice flown aboard a space shuttle mission. Structural synaptic plasticity was detected in the medial extrastriola, a region associated with encoding static head position, as decreased synapse density. These results are remarkably congruent with a recent report of decreased utricular function in astronauts immediately after returning from the International Space Station.
Collapse
Affiliation(s)
- David R Sultemeier
- Department of Head & Neck Surgery, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Kristel R Choy
- Department of Head & Neck Surgery, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Felix E Schweizer
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, California; and.,Brain Research Institute, David Geffen School of Medicine, University of California, Los Angeles, California
| | - Larry F Hoffman
- Department of Head & Neck Surgery, David Geffen School of Medicine, University of California, Los Angeles, California; .,Brain Research Institute, David Geffen School of Medicine, University of California, Los Angeles, California
| |
Collapse
|