1
|
Ono K, Jarysta A, Hughes NC, Jukic A, Chang HHV, Deans MR, Eatock RA, Cullen KE, Kindt KS, Tarchini B. Contributions of mirror-image hair cell orientation to mouse otolith organ and zebrafish neuromast function. eLife 2024; 13:RP97674. [PMID: 39531034 PMCID: PMC11556791 DOI: 10.7554/elife.97674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Otolith organs in the inner ear and neuromasts in the fish lateral-line harbor two populations of hair cells oriented to detect stimuli in opposing directions. The underlying mechanism is highly conserved: the transcription factor EMX2 is regionally expressed in just one hair cell population and acts through the receptor GPR156 to reverse cell orientation relative to the other population. In mouse and zebrafish, loss of Emx2 results in sensory organs that harbor only one hair cell orientation and are not innervated properly. In zebrafish, Emx2 also confers hair cells with reduced mechanosensory properties. Here, we leverage mouse and zebrafish models lacking GPR156 to determine how detecting stimuli of opposing directions serves vestibular function, and whether GPR156 has other roles besides orienting hair cells. We find that otolith organs in Gpr156 mouse mutants have normal zonal organization and normal type I-II hair cell distribution and mechano-electrical transduction properties. In contrast, gpr156 zebrafish mutants lack the smaller mechanically evoked signals that characterize Emx2-positive hair cells. Loss of GPR156 does not affect orientation-selectivity of afferents in mouse utricle or zebrafish neuromasts. Consistent with normal otolith organ anatomy and afferent selectivity, Gpr156 mutant mice do not show overt vestibular dysfunction. Instead, performance on two tests that engage otolith organs is significantly altered - swimming and off-vertical-axis rotation. We conclude that GPR156 relays hair cell orientation and transduction information downstream of EMX2, but not selectivity for direction-specific afferents. These results clarify how molecular mechanisms that confer bi-directionality to sensory organs contribute to function, from single hair cell physiology to animal behavior.
Collapse
Affiliation(s)
- Kazuya Ono
- Department of Neurobiology, University of ChicagoChicagoUnited States
| | | | - Natasha C Hughes
- Department of Biomedical Engineering, Johns Hopkins UniversityBaltimoreUnited States
| | - Alma Jukic
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | - Hui Ho Vanessa Chang
- Department of Biomedical Engineering, Johns Hopkins UniversityBaltimoreUnited States
| | - Michael R Deans
- Department of Neurobiology, Spencer Fox Eccles School of Medicine, University of UtahSalt Lake CityUnited States
- Department of Otolaryngology - Head & Neck Surgery, Spencer Fox Eccles School of Medicine at the University of UtahSalt Lake CityUnited States
| | - Ruth Anne Eatock
- Department of Neurobiology, University of ChicagoChicagoUnited States
| | - Kathleen E Cullen
- Department of Biomedical Engineering, Johns Hopkins UniversityBaltimoreUnited States
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins UniversityBaltimoreUnited States
- Department of Neuroscience, Johns Hopkins UniversityBaltimoreUnited States
- Kavli Neuroscience Discovery Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Katie S Kindt
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | - Basile Tarchini
- The Jackson LaboratoryBar HarborUnited States
- Tufts University School of MedicineBostonUnited States
| |
Collapse
|
2
|
David S, Pinter K, Nguyen KK, Lee DS, Lei Z, Sokolova Y, Sheets L, Kindt KS. Kif1a and intact microtubules maintain synaptic-vesicle populations at ribbon synapses in zebrafish hair cells. J Physiol 2024:10.1113/JP286263. [PMID: 39373584 PMCID: PMC11973241 DOI: 10.1113/jp286263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/05/2024] [Indexed: 10/08/2024] Open
Abstract
Sensory hair cells of the inner ear utilize specialized ribbon synapses to transmit sensory stimuli to the central nervous system. This transmission necessitates rapid and sustained neurotransmitter release, which depends on a large pool of synaptic vesicles at the hair-cell presynapse. While previous work in neurons has shown that kinesin motor proteins traffic synaptic material along microtubules to the presynapse, the mechanisms of this process in hair cells remain unclear. Our study demonstrates that the kinesin motor protein Kif1a, along with an intact microtubule network, is essential for enriching synaptic vesicles at the presynapse in hair cells. Through genetic and pharmacological approaches, we disrupt Kif1a function and impair microtubule networks in hair cells of the zebrafish lateral-line system. These manipulations led to a significant reduction in synaptic-vesicle populations at the presynapse in hair cells. Using electron microscopy, in vivo calcium imaging, and electrophysiology, we show that a diminished supply of synaptic vesicles adversely affects ribbon-synapse function. Kif1aa mutants exhibit dramatic reductions in spontaneous vesicle release and evoked postsynaptic calcium responses. Furthermore, kif1aa mutants exhibit impaired rheotaxis, a behaviour reliant on the ability of hair cells in the lateral line to respond to sustained flow stimuli. Overall, our results demonstrate that Kif1a-mediated microtubule transport is critical to enrich synaptic vesicles at the active zone, a process that is vital for proper ribbon-synapse function in hair cells. KEY POINTS: Kif1a mRNAs are present in zebrafish hair cells. Loss of Kif1a disrupts the enrichment of synaptic vesicles at ribbon synapses. Disruption of microtubules depletes synaptic vesicles at ribbon synapses. Kif1aa mutants have impaired ribbon-synapse and sensory-system function.
Collapse
Affiliation(s)
- Sandeep David
- Section on Sensory Cell Development and Function, National Institute on Deafness and other Communication Disorders, Bethesda, Maryland, USA
- National Institutes of Health-Brown University Graduate Partnership Program, Bethesda, Maryland, USA
| | - Katherine Pinter
- Section on Sensory Cell Development and Function, National Institute on Deafness and other Communication Disorders, Bethesda, Maryland, USA
| | - Keziah-Khue Nguyen
- Department of Otolaryngology, Head and Neck Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - David S Lee
- Department of Otolaryngology, Head and Neck Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Zhengchang Lei
- Section on Sensory Cell Development and Function, National Institute on Deafness and other Communication Disorders, Bethesda, Maryland, USA
| | - Yuliya Sokolova
- Advanced Imaging Core, National Institute on Deafness and other Communication Disorders, Bethesda, Maryland, USA
| | - Lavinia Sheets
- Department of Otolaryngology, Head and Neck Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Katie S Kindt
- Section on Sensory Cell Development and Function, National Institute on Deafness and other Communication Disorders, Bethesda, Maryland, USA
| |
Collapse
|
3
|
Jukic A, Lei Z, Cebul ER, Pinter K, Tadesse Y, Jarysta A, David S, Mosqueda N, Tarchini B, Kindt K. Presynaptic Nrxn3 is essential for ribbon-synapse maturation in hair cells. Development 2024; 151:dev202723. [PMID: 39254120 PMCID: PMC11488651 DOI: 10.1242/dev.202723] [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: 01/25/2024] [Accepted: 08/28/2024] [Indexed: 09/11/2024]
Abstract
Hair cells of the inner ear and lateral-line system rely on specialized ribbon synapses to transmit sensory information to the central nervous system. The molecules required to assemble these synapses are not fully understood. We show that Nrxn3, a presynaptic adhesion molecule, is crucial for ribbon-synapse maturation in hair cells. In both mouse and zebrafish models, the loss of Nrxn3 results in significantly fewer intact ribbon synapses. We show in zebrafish that, initially, Nrxn3 loss does not alter pre- and postsynapse numbers but, later, synapses fail to pair, leading to postsynapse loss. We also demonstrate that Nrxn3 subtly influences synapse selectivity in zebrafish lateral-line hair cells that detect anterior flow. Loss of Nrxn3 leads to a 60% loss of synapses in zebrafish, which dramatically reduces pre- and postsynaptic responses. Despite fewer synapses, auditory responses in zebrafish and mice are unaffected. This work demonstrates that Nrxn3 is a crucial and conserved molecule required for the maturation of ribbon synapses. Understanding how ribbon synapses mature is essential to generating new therapies to treat synaptopathies linked to auditory or vestibular dysfunction.
Collapse
Affiliation(s)
- Alma Jukic
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - Zhengchang Lei
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - Elizabeth R. Cebul
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - Katherine Pinter
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - Yommi Tadesse
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | | | - Sandeep David
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - Natalie Mosqueda
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| | - Basile Tarchini
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Katie Kindt
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, Bethesda, MD 20892, USA
| |
Collapse
|
4
|
Ono K, Jarysta A, Hughes NC, Jukic A, Chang HHV, Deans MR, Eatock RA, Cullen KE, Kindt K, Tarchini B. Contributions of mirror-image hair cell orientation to mouse otolith organ and zebrafish neuromast function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.26.586740. [PMID: 39282410 PMCID: PMC11398332 DOI: 10.1101/2024.03.26.586740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
Otolith organs in the inner ear and neuromasts in the fish lateral-line harbor two populations of hair cells oriented to detect stimuli in opposing directions. The underlying mechanism is highly conserved: the transcription factor EMX2 is regionally expressed in just one hair cell population and acts through the receptor GPR156 to reverse cell orientation relative to the other population. In mouse and zebrafish, loss of Emx2 results in sensory organs that harbor only one hair cell orientation and are not innervated properly. In zebrafish, Emx2 also confers hair cells with reduced mechanosensory properties. Here, we leverage mouse and zebrafish models lacking GPR156 to determine how detecting stimuli of opposing directions serves vestibular function, and whether GPR156 has other roles besides orienting hair cells. We find that otolith organs in Gpr156 mouse mutants have normal zonal organization and normal type I-II hair cell distribution and mechano-electrical transduction properties. In contrast, gpr156 zebrafish mutants lack the smaller mechanically-evoked signals that characterize Emx2-positive hair cells. Loss of GPR156 does not affect orientation-selectivity of afferents in mouse utricle or zebrafish neuromasts. Consistent with normal otolith organ anatomy and afferent selectivity, Gpr156 mutant mice do not show overt vestibular dysfunction. Instead, performance on two tests that engage otolith organs is significantly altered - swimming and off-vertical-axis rotation. We conclude that GPR156 relays hair cell orientation and transduction information downstream of EMX2, but not selectivity for direction-specific afferents. These results clarify how molecular mechanisms that confer bi-directionality to sensory organs contribute to function, from single hair cell physiology to animal behavior.
Collapse
Affiliation(s)
- Kazuya Ono
- Department of Neurobiology, University of Chicago, Chicago, IL, 60637, USA
| | | | - Natasha C Hughes
- Dept. of Biomedical Engineering, Johns Hopkins University, Baltimore, 21205 MD, USA
| | - Alma Jukic
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MA, USA
| | - Hui Ho Vanessa Chang
- Dept. of Biomedical Engineering, Johns Hopkins University, Baltimore, 21205 MD, USA
| | - Michael R Deans
- Department of Neurobiology, Spencer Fox Eccles School of Medicine, University of Utah, Salt Lake City, Utah, USA
- Department of Otolaryngology - Head & Neck Surgery, Spencer Fox Eccles School of Medicine at the University of Utah, Salt Lake City, Utah, USA
| | - Ruth Anne Eatock
- Department of Neurobiology, University of Chicago, Chicago, IL, 60637, USA
| | - Kathleen E Cullen
- Dept. of Biomedical Engineering, Johns Hopkins University, Baltimore, 21205 MD, USA
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University, Baltimore 21205 MD, USA
- Department of Neuroscience, Johns Hopkins University, Baltimore 21205 MD, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore 21205 MD, USA
| | - Katie Kindt
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MA, USA
| | - Basile Tarchini
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
- Tufts University School of Medicine, Boston, MA 02111, USA
| |
Collapse
|
5
|
David S, Pinter K, Nguyen KK, Lee DS, Lei Z, Sokolova Y, Sheets L, Kindt KS. Kif1a and intact microtubules maintain synaptic-vesicle populations at ribbon synapses in zebrafish hair cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.20.595037. [PMID: 38903095 PMCID: PMC11188139 DOI: 10.1101/2024.05.20.595037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Sensory hair cells of the inner ear utilize specialized ribbon synapses to transmit sensory stimuli to the central nervous system. This sensory transmission necessitates rapid and sustained neurotransmitter release, which relies on a large pool of synaptic vesicles at the hair-cell presynapse. Work in neurons has shown that kinesin motor proteins traffic synaptic material along microtubules to the presynapse, but how new synaptic material reaches the presynapse in hair cells is not known. We show that the kinesin motor protein Kif1a and an intact microtubule network are necessary to enrich synaptic vesicles at the presynapse in hair cells. We use genetics and pharmacology to disrupt Kif1a function and impair microtubule networks in hair cells of the zebrafish lateral-line system. We find that these manipulations decrease synaptic-vesicle populations at the presynapse in hair cells. Using electron microscopy, along with in vivo calcium imaging and electrophysiology, we show that a diminished supply of synaptic vesicles adversely affects ribbon-synapse function. Kif1a mutants exhibit dramatic reductions in spontaneous vesicle release and evoked postsynaptic calcium responses. Additionally, we find that kif1a mutants exhibit impaired rheotaxis, a behavior reliant on the ability of hair cells in the lateral line to respond to sustained flow stimuli. Overall, our results demonstrate that Kif1a-based microtubule transport is critical to enrich synaptic vesicles at the active zone in hair cells, a process that is vital for proper ribbon-synapse function.
Collapse
Affiliation(s)
- Sandeep David
- Section on Sensory Cell Development and Function, National Institute on Deafness and other Communication Disorders, Bethesda, MD, USA
- National Institutes of Health-Brown University Graduate Partnership Program, Bethesda, MD, USA
| | - Katherine Pinter
- Section on Sensory Cell Development and Function, National Institute on Deafness and other Communication Disorders, Bethesda, MD, USA
| | - Keziah-Khue Nguyen
- Department of Otolaryngology - Head and Neck Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - David S Lee
- Department of Otolaryngology - Head and Neck Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Zhengchang Lei
- Section on Sensory Cell Development and Function, National Institute on Deafness and other Communication Disorders, Bethesda, MD, USA
| | - Yuliya Sokolova
- Advanced Imaging Core, National Institute on Deafness and other Communication Disorders, Bethesda, MD, USA
| | - Lavinia Sheets
- Department of Otolaryngology - Head and Neck Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Katie S Kindt
- Section on Sensory Cell Development and Function, National Institute on Deafness and other Communication Disorders, Bethesda, MD, USA
| |
Collapse
|
6
|
Suppermpool A, Lyons DG, Broom E, Rihel J. Sleep pressure modulates single-neuron synapse number in zebrafish. Nature 2024; 629:639-645. [PMID: 38693264 PMCID: PMC11096099 DOI: 10.1038/s41586-024-07367-3] [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: 11/14/2022] [Accepted: 03/27/2024] [Indexed: 05/03/2024]
Abstract
Sleep is a nearly universal behaviour with unclear functions1. The synaptic homeostasis hypothesis proposes that sleep is required to renormalize the increases in synaptic number and strength that occur during wakefulness2. Some studies examining either large neuronal populations3 or small patches of dendrites4 have found evidence consistent with the synaptic homeostasis hypothesis, but whether sleep merely functions as a permissive state or actively promotes synaptic downregulation at the scale of whole neurons is unclear. Here, by repeatedly imaging all excitatory synapses on single neurons across sleep-wake states of zebrafish larvae, we show that synapses are gained during periods of wake (either spontaneous or forced) and lost during sleep in a neuron-subtype-dependent manner. However, synapse loss is greatest during sleep associated with high sleep pressure after prolonged wakefulness, and lowest in the latter half of an undisrupted night. Conversely, sleep induced pharmacologically during periods of low sleep pressure is insufficient to trigger synapse loss unless adenosine levels are boosted while noradrenergic tone is inhibited. We conclude that sleep-dependent synapse loss is regulated by sleep pressure at the level of the single neuron and that not all sleep periods are equally capable of fulfilling the functions of synaptic homeostasis.
Collapse
Affiliation(s)
- Anya Suppermpool
- Department of Cell and Developmental Biology, University College London, London, UK
- UCL Ear Institute, University College London, London, UK
| | - Declan G Lyons
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Elizabeth Broom
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Jason Rihel
- Department of Cell and Developmental Biology, University College London, London, UK.
| |
Collapse
|
7
|
Li J, Miramontes TG, Czopka T, Monk KR. Synaptic input and Ca 2+ activity in zebrafish oligodendrocyte precursor cells contribute to myelin sheath formation. Nat Neurosci 2024; 27:219-231. [PMID: 38216650 DOI: 10.1038/s41593-023-01553-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 12/13/2023] [Indexed: 01/14/2024]
Abstract
In the nervous system, only one type of neuron-glial synapse is known to exist: that between neurons and oligodendrocyte precursor cells (OPCs), yet their composition, assembly, downstream signaling and in vivo functions remain largely unclear. Here, we address these questions using in vivo microscopy in zebrafish spinal cord and identify postsynaptic molecules PSD-95 and gephyrin in OPCs. The puncta containing these molecules in OPCs increase during early development and decrease upon OPC differentiation. These puncta are highly dynamic and frequently assemble at 'hotspots'. Gephyrin hotspots and synapse-associated Ca2+ activity in OPCs predict where a subset of myelin sheaths forms in differentiated oligodendrocytes. Further analyses reveal that spontaneous synaptic release is integral to OPC Ca2+ activity, while evoked synaptic release contributes only in early development. Finally, disruption of the synaptic genes dlg4a/dlg4b, gphnb and nlgn3b impairs OPC differentiation and myelination. Together, we propose that neuron-OPC synapses are dynamically assembled and can predetermine myelination patterns through Ca2+ signaling.
Collapse
Affiliation(s)
- Jiaxing Li
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA.
| | | | - Tim Czopka
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Kelly R Monk
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
8
|
Zhu J, Lv C, Henry D, Viviano S, Santos-Sacchi J, Matthews G, Zenisek D. Role of Ribeye PXDLS/T-binding cleft in normal synaptic ribbon function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.12.571266. [PMID: 38168344 PMCID: PMC10760060 DOI: 10.1101/2023.12.12.571266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Non-spiking sensory hair cells of the auditory and vestibular systems encode a dynamic range of graded signals with high fidelity by vesicle exocytosis at ribbon synapses. Ribeye, the most abundant protein in the synaptic ribbon, is composed of a unique A domain specific for ribbons and a B-domain nearly identical to the transcriptional corepressor CtBP2. CTBP2 and the B-domain of Ribeye contain a surface cleft that binds to proteins harboring a PXDLS/T peptide motif. Little is known about the importance of this binding site in synaptic function. Piccolo has a well-conserved PVDLT motif and we find that overexpressed Ribeye exhibits striking co-localization with Piccolo in INS-cells, while two separate mutants containing mutations in PXDLS/T-binding region, fail to co-localize with Piccolo. Similarly, co-transfected Ribeye and a piccolo fragment containing the PVDLT region co-localize in HEK cells. Expression of wild-type Ribeye-YFP in zebrafish neuromast hair cells returns electron densities to ribbon structures and mostly rescued normal synaptic transmission and morphological phenotypes in a mutant zebrafish lacking most Ribeye. By contrast, Ribeye-YFP harboring a mutation in the PXDLS/T-binding cleft resulted in ectopic electron dense aggregates that did not collect vesicles and the persistence of ribbons lacking electron densities. Furthermore, overexpression failed to return capacitance responses to normal levels. These results point toward a role for the PXDLS/T-binding cleft in the recruitment of Ribeye to ribbons and in normal synaptic function.
Collapse
Affiliation(s)
- Jie Zhu
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520
| | - Caixia Lv
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520
| | - Diane Henry
- Program in Neuroscience, State University of New York, Stony Brook, New York 11759
| | - Stephen Viviano
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520
| | - Joseph Santos-Sacchi
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520
- Neuroscience, Yale University School of Medicine, New Haven, CT 06520
- Surgery (Otolaryngology), Yale University School of Medicine, New Haven, CT 06520
| | - Gary Matthews
- Program in Neuroscience, State University of New York, Stony Brook, New York 11759
| | - David Zenisek
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520
- Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT 06520
- Neuroscience, Yale University School of Medicine, New Haven, CT 06520
| |
Collapse
|
9
|
Shrestha AP, Rameshkumar N, Boff JM, Rajmanna R, Chandrasegaran T, Frederick CE, Zenisek D, Vaithianathan T. The Effects of Aging on Rod Bipolar Cell Ribbon Synapses. Cells 2023; 12:2385. [PMID: 37830599 PMCID: PMC10572008 DOI: 10.3390/cells12192385] [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: 08/20/2023] [Revised: 09/20/2023] [Accepted: 09/27/2023] [Indexed: 10/14/2023] Open
Abstract
The global health concern posed by age-related visual impairment highlights the need for further research focused on the visual changes that occur during the process of aging. To date, multiple sensory alterations related to aging have been identified, including morphological and functional changes in inner hair cochlear cells, photoreceptors, and retinal ganglion cells. While some age-related morphological changes are known to occur in rod bipolar cells in the retina, their effects on these cells and on their connection to other cells via ribbon synapses remain elusive. To investigate the effects of aging on rod bipolar cells and their ribbon synapses, we compared synaptic calcium currents, calcium dynamics, and exocytosis in zebrafish (Danio rerio) that were middle-aged (MA,18 months) or old-aged (OA, 36 months). The bipolar cell terminal in OA zebrafish exhibited a two-fold reduction in number of synaptic ribbons, an increased ribbon length, and a decrease in local Ca2+ signals at the tested ribbon location, with little change in the overall magnitude of the calcium current or exocytosis in response to brief pulses. Staining of the synaptic ribbons with antibodies specific for PKCa revealed shortening of the inner nuclear and plexiform layers (INL and IPL). These findings shed light on age-related changes in the retina that are related to synaptic ribbons and calcium signals.
Collapse
Affiliation(s)
- Abhishek P. Shrestha
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Nirujan Rameshkumar
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Johane M. Boff
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Rhea Rajmanna
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | | | - Courtney E. Frederick
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA (D.Z.)
| | - David Zenisek
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA (D.Z.)
| | - Thirumalini Vaithianathan
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Department of Ophthalmology, Hamilton Eye Institute, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
10
|
Baeza-Loya S, Raible DW. Vestibular physiology and function in zebrafish. Front Cell Dev Biol 2023; 11:1172933. [PMID: 37143895 PMCID: PMC10151581 DOI: 10.3389/fcell.2023.1172933] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/06/2023] [Indexed: 05/06/2023] Open
Abstract
The vestibular system of the inner ear provides information about head motion and spatial orientation relative to gravity to ensure gaze stability, balance, and postural control. Zebrafish, like humans, have five sensory patches per ear that serve as peripheral vestibular organs, with the addition of the lagena and macula neglecta. The zebrafish inner ear can be easily studied due to its accessible location, the transparent tissue of larval fish, and the early development of vestibular behaviors. Thus, zebrafish are an excellent model for studying the development, physiology, and function of the vestibular system. Recent work has made great strides to elucidate vestibular neural circuitry in fish, tracing sensory transmission from receptors in the periphery to central computational circuits driving vestibular reflexes. Here we highlight recent work that illuminates the functional organization of vestibular sensory epithelia, innervating first-order afferent neurons, and second-order neuronal targets in the hindbrain. Using a combination of genetic, anatomical, electrophysiological, and optical techniques, these studies have probed the roles of vestibular sensory signals in fish gaze, postural, and swimming behaviors. We discuss remaining questions in vestibular development and organization that are tractable in the zebrafish model.
Collapse
Affiliation(s)
| | - David W. Raible
- Virginia Merrill Bloedel Hearing Research Center, Department of Otolaryngology-HNS and Biological Structure, University of Washington, Seattle, WA, United States
| |
Collapse
|
11
|
Riley KC, Koleilat A, Dugdale JA, Cooper SA, Christensen TA, Schimmenti LA. Three-Dimensional Structure of Inner Ear Hair Cell Ribbon Synapses in a Zebrafish Model of Usher Syndrome Type 1B. Zebrafish 2023; 20:47-54. [PMID: 37071854 DOI: 10.1089/zeb.2022.0049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023] Open
Abstract
Our understanding of inner ear hair cell ultrastructure has heretofore relied upon two-dimensional imaging; however, serial block-face scanning electron microscopy (SBFSEM) changes this paradigm allowing for three-dimensional evaluation. We compared inner ear hair cells of the apical cristae in myo7aa-/- null zebrafish, a model of human Usher Syndrome type 1B, to hair cells in wild-type zebrafish by SBFSEM to investigate possible ribbon synapse ultrastructural differences. Previously, it has been shown that compared to wild type, myo7aa-/- zebrafish neuromast hair cells have fewer ribbon synapses yet similar ribbon areas. We expect the recapitulation of these results within the inner ear apical crista hair cells furthering the knowledge of three-dimensional ribbon synapse structure while resolving the feasibility of therapeutically targeting myo7aa-/- mutant ribbons. In this report, we evaluated ribbon synapse number, volume, surface area, and sphericity. Localization of ribbons and their distance from the nearest innervation were also evaluated. We determined that myo7aa-/- mutant ribbon synapses are smaller in volume and surface area; however, all other measurements were not significantly different from wild-type zebrafish. Because the ribbon synapses are nearly indistinguishable between the myo7aa-/- mutant and wild type, it suggests that the ribbons are structurally receptive, supporting that therapeutic intervention may be feasible.
Collapse
Affiliation(s)
- Kenneth C Riley
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota, USA
| | - Alaa Koleilat
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Joseph A Dugdale
- Department of Otorhinolaryngology, Head and Neck Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Shawna A Cooper
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Trace A Christensen
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
- Microscopy and Cell Analysis Core, and Mayo Clinic, Rochester, Minnesota, USA
| | - Lisa A Schimmenti
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota, USA
- Department of Otorhinolaryngology, Head and Neck Surgery, Mayo Clinic, Rochester, Minnesota, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
12
|
Iyer AA, Hosamani I, Nguyen JD, Cai T, Singh S, McGovern MM, Beyer L, Zhang H, Jen HI, Yousaf R, Birol O, Sun JJ, Ray RS, Raphael Y, Segil N, Groves AK. Cellular reprogramming with ATOH1, GFI1, and POU4F3 implicate epigenetic changes and cell-cell signaling as obstacles to hair cell regeneration in mature mammals. eLife 2022; 11:e79712. [PMID: 36445327 PMCID: PMC9708077 DOI: 10.7554/elife.79712] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 11/16/2022] [Indexed: 11/30/2022] Open
Abstract
Reprogramming of the cochlea with hair-cell-specific transcription factors such as ATOH1 has been proposed as a potential therapeutic strategy for hearing loss. ATOH1 expression in the developing cochlea can efficiently induce hair cell regeneration but the efficiency of hair cell reprogramming declines rapidly as the cochlea matures. We developed Cre-inducible mice to compare hair cell reprogramming with ATOH1 alone or in combination with two other hair cell transcription factors, GFI1 and POU4F3. In newborn mice, all transcription factor combinations tested produced large numbers of cells with the morphology of hair cells and rudimentary mechanotransduction properties. However, 1 week later, only a combination of ATOH1, GFI1 and POU4F3 could reprogram non-sensory cells of the cochlea to a hair cell fate, and these new cells were less mature than cells generated by reprogramming 1 week earlier. We used scRNA-seq and combined scRNA-seq and ATAC-seq to suggest at least two impediments to hair cell reprogramming in older animals. First, hair cell gene loci become less epigenetically accessible in non-sensory cells of the cochlea with increasing age. Second, signaling from hair cells to supporting cells, including Notch signaling, can prevent reprogramming of many supporting cells to hair cells, even with three hair cell transcription factors. Our results shed light on the molecular barriers that must be overcome to promote hair cell regeneration in the adult cochlea.
Collapse
Affiliation(s)
- Amrita A Iyer
- Department of Molecular & Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Ishwar Hosamani
- Department of Molecular & Human Genetics, Baylor College of MedicineHoustonUnited States
| | - John D Nguyen
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology at USCLos AngelesUnited States
| | - Tiantian Cai
- Program in Developmental Biology, Baylor College of MedicineHoustonUnited States
| | - Sunita Singh
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Melissa M McGovern
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Lisa Beyer
- Department of Otolaryngology-Head and Neck Surgery, University of MichiganAnn ArborUnited States
| | - Hongyuan Zhang
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Hsin-I Jen
- Program in Developmental Biology, Baylor College of MedicineHoustonUnited States
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Rizwan Yousaf
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Onur Birol
- Program in Developmental Biology, Baylor College of MedicineHoustonUnited States
| | - Jenny J Sun
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Russell S Ray
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Yehoash Raphael
- Department of Otolaryngology-Head and Neck Surgery, University of MichiganAnn ArborUnited States
| | - Neil Segil
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of the University of Southern California, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology at USCLos AngelesUnited States
- Caruso Department of Otolaryngology-Head and Neck Surgery, Keck School of Medicine of the University of Southern CaliforniaLos AngelesUnited States
| | - Andrew K Groves
- Department of Molecular & Human Genetics, Baylor College of MedicineHoustonUnited States
- Program in Developmental Biology, Baylor College of MedicineHoustonUnited States
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
13
|
Shrestha AP, Saravanakumar A, Konadu B, Madireddy S, Gibert Y, Vaithianathan T. Embryonic Hyperglycemia Delays the Development of Retinal Synapses in a Zebrafish Model. Int J Mol Sci 2022; 23:ijms23179693. [PMID: 36077087 PMCID: PMC9456524 DOI: 10.3390/ijms23179693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/16/2022] [Accepted: 08/19/2022] [Indexed: 11/22/2022] Open
Abstract
Embryonic hyperglycemia negatively impacts retinal development, leading to abnormal visual behavior, altered timing of retinal progenitor differentiation, decreased numbers of retinal ganglion cells and Müller glia, and vascular leakage. Because synaptic disorganization is a prominent feature of many neurological diseases, the goal of the current work was to study the potential impact of hyperglycemia on retinal ribbon synapses during embryonic development. Our approach utilized reverse transcription quantitative PCR (RT-qPCR) and immunofluorescence labeling to compare the transcription of synaptic proteins and their localization in hyperglycemic zebrafish embryos, respectively. Our data revealed that the maturity of synaptic ribbons was compromised in hyperglycemic zebrafish larvae, where altered ribeye expression coincided with the delay in establishing retinal ribbon synapses and an increase in the immature synaptic ribbons. Our results suggested that embryonic hyperglycemia disrupts retinal synapses by altering the development of the synaptic ribbon, which can lead to visual defects. Future studies using zebrafish models of hyperglycemia will allow us to study the underlying mechanisms of retinal synapse development.
Collapse
Affiliation(s)
- Abhishek P. Shrestha
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Ambalavanan Saravanakumar
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Program in Biology, Rhodes College, Memphis, TN 38112, USA
| | - Bridget Konadu
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Saivikram Madireddy
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Yann Gibert
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Thirumalini Vaithianathan
- Department of Pharmacology, Addiction Science, and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Correspondence: ; Tel.: +1-901-448-2786
| |
Collapse
|
14
|
Shankhwar S, Schwarz K, Katiyar R, Jung M, Maxeiner S, Südhof TC, Schmitz F. RIBEYE B-Domain Is Essential for RIBEYE A-Domain Stability and Assembly of Synaptic Ribbons. Front Mol Neurosci 2022; 15:838311. [PMID: 35153673 PMCID: PMC8831697 DOI: 10.3389/fnmol.2022.838311] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/03/2022] [Indexed: 01/17/2023] Open
Abstract
Synaptic ribbons are presynaptic specializations that define eponymous ribbon synapses. Synaptic ribbons are largely composed of RIBEYE, a protein containing an N-terminal A-domain and a carboxyterminal B-domain that is identical with CtBP2, a NAD(H)-binding transcriptional co-repressor. Previously we showed that synaptic ribbons are completely absent in RIBEYE knockout mice in which the RIBEYE A-domain-encoding exon had been deleted, but CtBP2 is still made, demonstrating that the A-domain is required for synaptic ribbon assembly. In the present study, we asked whether the RIBEYE B-domain also has an essential role in the assembly of synaptic ribbons. For this purpose, we made use of RIBEYE knockin mice in which the RIBEYE B-domain was replaced by a fluorescent protein domain, whereas the RIBEYE A-domain was retained unchanged. We found that replacing the RIBEYE B-domain with a fluorescent protein module destabilizes the resulting hybrid protein and causes a complete loss of synaptic ribbons. Our results thus demonstrate an essential role of the RIBEYE B-domain in enabling RIBEYE assembly into synaptic ribbons, reinforcing the notion that RIBEYE is the central organizer of synaptic ribbons.
Collapse
Affiliation(s)
- Soni Shankhwar
- Institute of Anatomy and Cell Biology, Saarland University, Medical School, Homburg, Germany
- *Correspondence: Soni Shankhwar Frank Schmitz
| | - Karin Schwarz
- Institute of Anatomy and Cell Biology, Saarland University, Medical School, Homburg, Germany
| | - Rashmi Katiyar
- Institute of Anatomy and Cell Biology, Saarland University, Medical School, Homburg, Germany
| | - Martin Jung
- Institute of Medical Biochemistry and Molecular Biology, Saarland University, Medical School, Homburg, Germany
| | - Stephan Maxeiner
- Institute of Anatomy and Cell Biology, Saarland University, Medical School, Homburg, Germany
| | - Thomas C. Südhof
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, United States
| | - Frank Schmitz
- Institute of Anatomy and Cell Biology, Saarland University, Medical School, Homburg, Germany
- *Correspondence: Soni Shankhwar Frank Schmitz
| |
Collapse
|
15
|
Pereida-Jaramillo E, Gómez-González GB, Espino-Saldaña AE, Martínez-Torres A. Calcium Signaling in the Cerebellar Radial Glia and Its Association with Morphological Changes during Zebrafish Development. Int J Mol Sci 2021; 22:ijms222413509. [PMID: 34948305 PMCID: PMC8706707 DOI: 10.3390/ijms222413509] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/08/2021] [Accepted: 12/12/2021] [Indexed: 01/02/2023] Open
Abstract
Radial glial cells are a distinct non-neuronal cell type that, during development, span the entire width of the brain walls of the ventricular system. They play a central role in the origin and placement of neurons, since their processes form structural scaffolds that guide and facilitate neuronal migration. Furthermore, glutamatergic signaling in the radial glia of the adult cerebellum (i.e., Bergmann glia), is crucial for precise motor coordination. Radial glial cells exhibit spontaneous calcium activity and functional coupling spread calcium waves. However, the origin of calcium activity in relation to the ontogeny of cerebellar radial glia has not been widely explored, and many questions remain unanswered regarding the role of radial glia in brain development in health and disease. In this study we used a combination of whole mount immunofluorescence and calcium imaging in transgenic (gfap-GCaMP6s) zebrafish to determine how development of calcium activity is related to morphological changes of the cerebellum. We found that the morphological changes in cerebellar radial glia are quite dynamic; the cells are remarkably larger and more elaborate in their soma size, process length and numbers after 7 days post fertilization. Spontaneous calcium events were scarce during the first 3 days of development and calcium waves appeared on day 5, which is associated with the onset of more complex morphologies of radial glia. Blockage of gap junction coupling inhibited the propagation of calcium waves, but not basal local calcium activity. This work establishes crucial clues in radial glia organization, morphology and calcium signaling during development and provides insight into its role in complex behavioral paradigms.
Collapse
|
16
|
Plazas PV, Elgoyhen AB. The Cholinergic Lateral Line Efferent Synapse: Structural, Functional and Molecular Similarities With Those of the Cochlea. Front Cell Neurosci 2021; 15:765083. [PMID: 34712122 PMCID: PMC8545859 DOI: 10.3389/fncel.2021.765083] [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: 08/26/2021] [Accepted: 09/21/2021] [Indexed: 11/25/2022] Open
Abstract
Vertebrate hair cell (HC) systems are innervated by efferent fibers that modulate their response to external stimuli. In mammals, the best studied efferent-HC synapse, the cholinergic medial olivocochlear (MOC) efferent system, makes direct synaptic contacts with HCs. The net effect of MOC activity is to hyperpolarize HCs through the activation of α9α10 nicotinic cholinergic receptors (nAChRs) and the subsequent activation of Ca2+-dependent SK2 potassium channels. A serious obstacle in research on many mammalian sensory systems in their native context is that their constituent neurons are difficult to access even in newborn animals, hampering circuit observation, mapping, or controlled manipulation. By contrast, fishes and amphibians have a superficial and accessible mechanosensory system, the lateral line (LL), which circumvents many of these problems. LL responsiveness is modulated by efferent neurons which aid to distinguish between external and self-generated stimuli. One component of the LL efferent system is cholinergic and its activation inhibits LL afferent activity, similar to what has been described for MOC efferents. The zebrafish (Danio rerio) has emerged as a powerful model system for studying human hearing and balance disorders, since LL HC are structurally and functionally analogous to cochlear HCs, but are optically and pharmacologically accessible within an intact specimen. Complementing mammalian studies, zebrafish have been used to gain significant insights into many facets of HC biology, including mechanotransduction and synaptic physiology as well as mechanisms of both hereditary and acquired HC dysfunction. With the rise of the zebrafish LL as a model in which to study auditory system function and disease, there has been an increased interest in studying its efferent system and evaluate the similarity between mammalian and piscine efferent synapses. Advances derived from studies in zebrafish include understanding the effect of the LL efferent system on HC and afferent activity, and revealing that an α9-containing nAChR, functionally coupled to SK channels, operates at the LL efferent synapse. In this review, we discuss the tools and findings of these recent investigations into zebrafish efferent-HC synapse, their commonalities with the mammalian counterpart and discuss several emerging areas for future studies.
Collapse
Affiliation(s)
- Paola V Plazas
- Instituto de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ana Belén Elgoyhen
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| |
Collapse
|
17
|
Holmgren M, Ravicz ME, Hancock KE, Strelkova O, Kallogjeri D, Indzhykulian AA, Warchol ME, Sheets L. Mechanical overstimulation causes acute injury and synapse loss followed by fast recovery in lateral-line neuromasts of larval zebrafish. eLife 2021; 10:69264. [PMID: 34665127 PMCID: PMC8555980 DOI: 10.7554/elife.69264] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 10/18/2021] [Indexed: 12/14/2022] Open
Abstract
Excess noise damages sensory hair cells, resulting in loss of synaptic connections with auditory nerves and, in some cases, hair-cell death. The cellular mechanisms underlying mechanically induced hair-cell damage and subsequent repair are not completely understood. Hair cells in neuromasts of larval zebrafish are structurally and functionally comparable to mammalian hair cells but undergo robust regeneration following ototoxic damage. We therefore developed a model for mechanically induced hair-cell damage in this highly tractable system. Free swimming larvae exposed to strong water wave stimulus for 2 hr displayed mechanical injury to neuromasts, including afferent neurite retraction, damaged hair bundles, and reduced mechanotransduction. Synapse loss was observed in apparently intact exposed neuromasts, and this loss was exacerbated by inhibiting glutamate uptake. Mechanical damage also elicited an inflammatory response and macrophage recruitment. Remarkably, neuromast hair-cell morphology and mechanotransduction recovered within hours following exposure, suggesting severely damaged neuromasts undergo repair. Our results indicate functional changes and synapse loss in mechanically damaged lateral-line neuromasts that share key features of damage observed in noise-exposed mammalian ear. Yet, unlike the mammalian ear, mechanical damage to neuromasts is rapidly reversible.
Collapse
Affiliation(s)
- Melanie Holmgren
- Department of Otolaryngology, Washington University School of Medicine, St Louis, United States
| | - Michael E Ravicz
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, United States.,Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, United States
| | - Kenneth E Hancock
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, United States.,Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, United States
| | - Olga Strelkova
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, United States.,Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, United States
| | - Dorina Kallogjeri
- Department of Otolaryngology, Washington University School of Medicine, St Louis, United States
| | - Artur A Indzhykulian
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, United States.,Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, United States
| | - Mark E Warchol
- Department of Otolaryngology, Washington University School of Medicine, St Louis, United States.,Department of Neuroscience, Washington University School of Medicine, St Louis, United States
| | - Lavinia Sheets
- Department of Otolaryngology, Washington University School of Medicine, St Louis, United States.,Department of Developmental Biology, Washington University School of Medicine, St. Louis, United States
| |
Collapse
|
18
|
Holmgren M, Sheets L. Influence of Mpv17 on Hair-Cell Mitochondrial Homeostasis, Synapse Integrity, and Vulnerability to Damage in the Zebrafish Lateral Line. Front Cell Neurosci 2021; 15:693375. [PMID: 34413725 PMCID: PMC8369198 DOI: 10.3389/fncel.2021.693375] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 06/16/2021] [Indexed: 01/27/2023] Open
Abstract
Noise exposure is particularly stressful to hair-cell mitochondria, which must produce enough energy to meet high metabolic demands as well as regulate local intracellular Ca2+ concentrations. Mitochondrial Inner Membrane Protein 17 (Mpv17) functions as a non-selective cation channel and plays a role in maintaining mitochondrial homeostasis. In zebrafish, hair cells in mpv17a9/a9 mutants displayed elevated levels of reactive oxygen species (ROS), elevated mitochondrial calcium, hyperpolarized transmembrane potential, and greater vulnerability to neomycin, indicating impaired mitochondrial function. Using a strong water current to overstimulate hair cells in the zebrafish lateral line, we observed mpv17a9/a9 mutant hair cells were more vulnerable to morphological disruption than wild type (WT) siblings simultaneously exposed to the same stimulus. To determine the role of mitochondrial homeostasis on hair-cell synapse integrity, we surveyed synapse number in mpv17a9/a9 mutants and WT siblings as well as the sizes of presynaptic dense bodies (ribbons) and postsynaptic densities immediately following stimulus exposure. We observed mechanically injured mpv17a9/a9 neuromasts were not more vulnerable to synapse loss; they lost a similar number of synapses per hair cell relative to WT. Additionally, we quantified the size of hair cell pre- and postsynaptic structures following stimulation and observed significantly enlarged WT postsynaptic densities, yet relatively little change in the size of mpv17a9/a9 postsynaptic densities following stimulation. These results suggest chronically impaired hair-cell mitochondrial activity influences postsynaptic size under homeostatic conditions but does not exacerbate synapse loss following mechanical injury.
Collapse
Affiliation(s)
- Melanie Holmgren
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, United States
| | - Lavinia Sheets
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, United States
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
19
|
Colón-Cruz L, Rodriguez-Morales R, Santana-Cruz A, Cantres-Velez J, Torrado-Tapias A, Lin SJ, Yudowski G, Kensler R, Marie B, Burgess SM, Renaud O, Varshney GK, Behra M. Cnr2 Is Important for Ribbon Synapse Maturation and Function in Hair Cells and Photoreceptors. Front Mol Neurosci 2021; 14:624265. [PMID: 33958989 PMCID: PMC8093779 DOI: 10.3389/fnmol.2021.624265] [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: 10/31/2020] [Accepted: 02/24/2021] [Indexed: 02/04/2023] Open
Abstract
The role of the cannabinoid receptor 2 (CNR2) is still poorly described in sensory epithelia. We found strong cnr2 expression in hair cells (HCs) of the inner ear and the lateral line (LL), a superficial sensory structure in fish. Next, we demonstrated that sensory synapses in HCs were severely perturbed in larvae lacking cnr2. Appearance and distribution of presynaptic ribbons and calcium channels (Cav1.3) were profoundly altered in mutant animals. Clustering of membrane-associated guanylate kinase (MAGUK) in post-synaptic densities (PSDs) was also heavily affected, suggesting a role for cnr2 for maintaining the sensory synapse. Furthermore, vesicular trafficking in HCs was strongly perturbed suggesting a retrograde action of the endocannabinoid system (ECs) via cnr2 that was modulating HC mechanotransduction. We found similar perturbations in retinal ribbon synapses. Finally, we showed that larval swimming behaviors after sound and light stimulations were significantly different in mutant animals. Thus, we propose that cnr2 is critical for the processing of sensory information in the developing larva.
Collapse
Affiliation(s)
- Luis Colón-Cruz
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
| | - Roberto Rodriguez-Morales
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
| | - Alexis Santana-Cruz
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
| | - Juan Cantres-Velez
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
| | - Aranza Torrado-Tapias
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
| | - Sheng-Jia Lin
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Guillermo Yudowski
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico.,School of Medicine, Institute of Neurobiology, University of Puerto Rico, San Juan, Puerto Rico
| | - Robert Kensler
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
| | - Bruno Marie
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico.,School of Medicine, Institute of Neurobiology, University of Puerto Rico, San Juan, Puerto Rico
| | - Shawn M Burgess
- Developmental Genomics Section, Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| | - Olivier Renaud
- Cell and Tissue Imaging Facility (PICT-IBiSA, FranceBioImaging), Institut Curie, PSL Research University, U934/UMR3215, Paris, France
| | - Gaurav K Varshney
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States
| | - Martine Behra
- Department of Anatomy and Neurobiology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico
| |
Collapse
|
20
|
Holmgren M, Sheets L. Using the Zebrafish Lateral Line to Understand the Roles of Mitochondria in Sensorineural Hearing Loss. Front Cell Dev Biol 2021; 8:628712. [PMID: 33614633 PMCID: PMC7892962 DOI: 10.3389/fcell.2020.628712] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 12/23/2020] [Indexed: 01/05/2023] Open
Abstract
Hair cells are the mechanosensory receptors of the inner ear and can be damaged by noise, aging, and ototoxic drugs. This damage often results in permanent sensorineural hearing loss. Hair cells have high energy demands and rely on mitochondria to produce ATP as well as contribute to intracellular calcium homeostasis. In addition to generating ATP, mitochondria produce reactive oxygen species, which can lead to oxidative stress, and regulate cell death pathways. Zebrafish lateral-line hair cells are structurally and functionally analogous to cochlear hair cells but are optically and pharmacologically accessible within an intact specimen, making the zebrafish a good model in which to study hair-cell mitochondrial activity. Moreover, the ease of genetic manipulation of zebrafish embryos allows for the study of mutations implicated in human deafness, as well as the generation of transgenic models to visualize mitochondrial calcium transients and mitochondrial activity in live organisms. Studies of the zebrafish lateral line have shown that variations in mitochondrial activity can predict hair-cell susceptibility to damage by aminoglycosides or noise exposure. In addition, antioxidants have been shown to protect against noise trauma and ototoxic drug–induced hair-cell death. In this review, we discuss the tools and findings of recent investigations into zebrafish hair-cell mitochondria and their involvement in cellular processes, both under homeostatic conditions and in response to noise or ototoxic drugs. The zebrafish lateral line is a valuable model in which to study the roles of mitochondria in hair-cell pathologies and to develop therapeutic strategies to prevent sensorineural hearing loss in humans.
Collapse
Affiliation(s)
- Melanie Holmgren
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, United States
| | - Lavinia Sheets
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, United States.,Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
21
|
Banono NS, Gawel K, De Witte L, Esguerra CV. Zebrafish Larvae Carrying a Splice Variant Mutation in cacna1d: A New Model for Schizophrenia-Like Behaviours? Mol Neurobiol 2021; 58:877-894. [PMID: 33057948 PMCID: PMC7843589 DOI: 10.1007/s12035-020-02160-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 10/02/2020] [Indexed: 12/16/2022]
Abstract
Persons with certain single nucleotide polymorphisms (SNPs) in the CACNA1D gene (encoding voltage-gated calcium channel subunit alpha 1-D) have increased risk of developing neuropsychiatric disorders such as bipolar, schizophrenia and autism. The molecular consequences of SNPs on gene expression and protein function are not well understood. Thus, the use of animal models to determine genotype-phenotype correlations is critical to understanding disease pathogenesis. Here, we describe the behavioural changes in larval zebrafish carrying an essential splice site mutation (sa17298) in cacna1da. Heterozygous mutation resulted in 50% reduction of splice variants 201 and 202 (haploinsufficiency), while homozygosity increased transcript levels of variant 201 above wild type (WT; gain-of-function, GOF). Due to low homozygote viability, we focused primarily on performing the phenotypic analysis on heterozygotes. Indeed, cacna1dasa17298/WT larvae displayed hyperlocomotion-a behaviour characterised in zebrafish as a surrogate phenotype for epilepsy, anxiety or psychosis-like behaviour. Follow-up tests ruled out anxiety or seizures, however, as neither thigmotaxis defects nor epileptiform-like discharges in larval brains were observed. We therefore focused on testing for potential "psychosis-like" behaviour by assaying cacna1dasa17298/WT larval locomotor activity under constant light, during light-dark transition and in startle response to dark flashes. Furthermore, exposure of larvae to the antipsychotics, risperidone and haloperidol reversed cacna1da-induced hyperactivity to WT levels while valproate decreased but did not reverse hyperactivity. Together, these findings demonstrate that cacna1da haploinsufficiency induces behaviours in larval zebrafish analogous to those observed in rodent models of psychosis. Future studies on homozygous mutants will determine how cacna1d GOF alters behaviour in this context.
Collapse
Affiliation(s)
- Nancy Saana Banono
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway (NCMM), Faculty of Medicine, University of Oslo, Gaustadalléen 21, Forskningsparken, 0349, Oslo, Norway
| | - Kinga Gawel
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway (NCMM), Faculty of Medicine, University of Oslo, Gaustadalléen 21, Forskningsparken, 0349, Oslo, Norway
- Department of Experimental and Clinical Pharmacology, Medical University of Lublin, Jaczewskiego Str. 8b, 20-090, Lublin, Poland
| | - Linus De Witte
- Pharmaceutical and Biological Sciences, AP Hogeschool Antwerpen, Antwerp, Belgium
| | - Camila V Esguerra
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway (NCMM), Faculty of Medicine, University of Oslo, Gaustadalléen 21, Forskningsparken, 0349, Oslo, Norway.
- School of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Sem Sælandsvei 24, 0371, Oslo, Norway.
| |
Collapse
|
22
|
Zeng R, Brown AD, Rogers LS, Lawrence OT, Clark JI, Sisneros JA. Age-related loss of auditory sensitivity in the zebrafish (Danio rerio). Hear Res 2021; 403:108189. [PMID: 33556775 DOI: 10.1016/j.heares.2021.108189] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 01/10/2021] [Accepted: 01/22/2021] [Indexed: 11/27/2022]
Abstract
Age-related hearing loss (ARHL), also known as presbycusis, is a widespread and debilitating condition impacting many older adults. Conventionally, researchers utilize mammalian model systems or human cadaveric tissue to study ARHL pathology. Recently, the zebrafish has become an effective and tractable model system for a wide variety of genetic and environmental auditory insults, but little is known about the incidence or extent of ARHL in zebrafish and other non-mammalian models. Here, we evaluated whether zebrafish exhibit age-related loss in auditory sensitivity. The auditory sensitivity of adult wild-type zebrafish (AB/WIK strain) from three adult age subgroups (13-month, 20-month, and 37-month) was characterized using the auditory evoked potential (AEP) recording technique. AEPs were elicited using pure tone stimuli (115-4500 Hz) presented via an underwater loudspeaker and recorded using shielded subdermal metal electrodes. Based on measures of sound pressure and particle acceleration, the mean AEP thresholds of 37-month-old fish [mean sound pressure level (SPL) = 122.2 dB ± 2.2 dB SE re: 1 μPa; mean particle acceleration level (PAL) = -27.5 ± 2.3 dB SE re: 1 ms-2] were approximately 9 dB higher than that of 20-month-old fish [(mean SPL = 113.1 ± 2.7 dB SE re: 1 μPa; mean PAL = -37.2 ± 2.8 dB re: 1 ms-2; p = 0.007)] and 6 dB higher than that of 13-month-old fish [(mean SPL = 116.3 ± 2.5 dB SE re: 1 μPa; mean PAL = -34.1 ± 2.6 dB SE re: 1 ms-2; p = 0.052)]. Lowest AEP thresholds for all three age groups were generally between 800 Hz and 1850 Hz, with no evidence for frequency-specific age-related loss. Our results suggest that zebrafish undergo age-related loss in auditory sensitivity, but the form and magnitude of loss is markedly different than in mammals, including humans. Future work is needed to further describe the incidence and extent of ARHL across vertebrate groups and to determine which, if any, ARHL mechanisms may be conserved across vertebrates to support meaningful comparative/translational studies.
Collapse
Affiliation(s)
- Ruiyu Zeng
- Department of Psychology, University of Washington, 413 Guthrie Hall, Box 351525, Seattle, WA 98195, United States.
| | - Andrew D Brown
- Department of Speech and Hearing Sciences, University of Washington, Seattle, WA 98105, United States; Virginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, WA 98195, United States
| | - Loranzie S Rogers
- Department of Psychology, University of Washington, 413 Guthrie Hall, Box 351525, Seattle, WA 98195, United States
| | - Owen T Lawrence
- Department of Biological Structure, University of Washington, Seattle, 98195, United States
| | - John I Clark
- Department of Biological Structure, University of Washington, Seattle, 98195, United States; Department of Ophthalmology, University of Washington, Seattle, 98195, United States
| | - Joseph A Sisneros
- Department of Psychology, University of Washington, 413 Guthrie Hall, Box 351525, Seattle, WA 98195, United States; Virginia Merrill Bloedel Hearing Research Center, University of Washington, Seattle, WA 98195, United States; Department of Biology, University of Washington, Seattle, WA 98195, United States
| |
Collapse
|
23
|
Koleilat A, Dugdale JA, Christenson TA, Bellah JL, Lambert AM, Masino MA, Ekker SC, Schimmenti LA. L-type voltage-gated calcium channel agonists mitigate hearing loss and modify ribbon synapse morphology in the zebrafish model of Usher syndrome type 1. Dis Model Mech 2020; 13:dmm043885. [PMID: 33361086 PMCID: PMC7710014 DOI: 10.1242/dmm.043885] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 09/15/2020] [Indexed: 01/19/2023] Open
Abstract
The mariner (myo7aa-/- ) mutant is a zebrafish model for Usher syndrome type 1 (USH1). To further characterize hair cell synaptic elements in myo7aa-/- mutants, we focused on the ribbon synapse and evaluated ultrastructure, number and distribution of immunolabeled ribbons, and postsynaptic densities. By transmission electron microscopy, we determined that myo7aa-/- zebrafish have fewer glutamatergic vesicles tethered to ribbon synapses, yet maintain a comparable ribbon area. In myo7aa-/- hair cells, immunolocalization of Ctbp2 showed fewer ribbon-containing cells in total and an altered distribution of Ctbp2 puncta compared to wild-type hair cells. myo7aa-/- mutants have fewer postsynaptic densities - as assessed by MAGUK immunolabeling - compared to wild-type zebrafish. We quantified the circular swimming behavior of myo7aa-/- mutant fish and measured a greater turning angle (absolute smooth orientation). It has previously been shown that L-type voltage-gated calcium channels are necessary for ribbon localization and occurrence of postsynaptic density; thus, we hypothesized and observed that L-type voltage-gated calcium channel agonists change behavioral and synaptic phenotypes in myo7aa-/- mutants in a drug-specific manner. Our results indicate that treatment with L-type voltage-gated calcium channel agonists alter hair cell synaptic elements and improve behavioral phenotypes of myo7aa-/- mutants. Our data support that L-type voltage-gated calcium channel agonists induce morphological changes at the ribbon synapse - in both the number of tethered vesicles and regarding the distribution of Ctbp2 puncta - shift swimming behavior and improve acoustic startle response.
Collapse
Affiliation(s)
- Alaa Koleilat
- College of Continuing and Professional Studies, University of Minnesota, Minneapolis, MN 55108, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Clinical and Translational Science Track, Rochester, MN 55905, USA
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN 55905, USA
| | - Joseph A Dugdale
- Department of Otorhinolaryngology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Jeffrey L Bellah
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN 55905, USA
- Department of Genetics and Development, Columbia University, New York City, NY 10032, USA
| | - Aaron M Lambert
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Mark A Masino
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Stephen C Ekker
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Lisa A Schimmenti
- Department of Otorhinolaryngology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Ophthalmology and Visual Neuroscience, University of Minnesota, Minneapolis, MN 55454, USA
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
24
|
Voorn RA, Vogl C. Molecular Assembly and Structural Plasticity of Sensory Ribbon Synapses-A Presynaptic Perspective. Int J Mol Sci 2020; 21:E8758. [PMID: 33228215 PMCID: PMC7699581 DOI: 10.3390/ijms21228758] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/13/2022] Open
Abstract
In the mammalian cochlea, specialized ribbon-type synapses between sensory inner hair cells (IHCs) and postsynaptic spiral ganglion neurons ensure the temporal precision and indefatigability of synaptic sound encoding. These high-through-put synapses are presynaptically characterized by an electron-dense projection-the synaptic ribbon-which provides structural scaffolding and tethers a large pool of synaptic vesicles. While advances have been made in recent years in deciphering the molecular anatomy and function of these specialized active zones, the developmental assembly of this presynaptic interaction hub remains largely elusive. In this review, we discuss the dynamic nature of IHC (pre-) synaptogenesis and highlight molecular key players as well as the transport pathways underlying this process. Since developmental assembly appears to be a highly dynamic process, we further ask if this structural plasticity might be maintained into adulthood, how this may influence the functional properties of a given IHC synapse and how such plasticity could be regulated on the molecular level. To do so, we take a closer look at other ribbon-bearing systems, such as retinal photoreceptors and pinealocytes and aim to infer conserved mechanisms that may mediate these phenomena.
Collapse
MESH Headings
- Alcohol Oxidoreductases/genetics
- Alcohol Oxidoreductases/metabolism
- Animals
- Co-Repressor Proteins/genetics
- Co-Repressor Proteins/metabolism
- Cytoskeletal Proteins/genetics
- Cytoskeletal Proteins/metabolism
- Cytoskeleton/metabolism
- Cytoskeleton/ultrastructure
- Gene Expression Regulation, Developmental
- Hair Cells, Auditory, Inner/metabolism
- Hair Cells, Auditory, Inner/ultrastructure
- Hair Cells, Auditory, Outer/metabolism
- Hair Cells, Auditory, Outer/ultrastructure
- Hair Cells, Vestibular/metabolism
- Hair Cells, Vestibular/ultrastructure
- Mechanotransduction, Cellular
- Mice
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Neuronal Plasticity/genetics
- Neuropeptides/genetics
- Neuropeptides/metabolism
- Rats
- Synapses/metabolism
- Synapses/ultrastructure
- Synaptic Transmission/genetics
- Synaptic Vesicles/metabolism
- Synaptic Vesicles/ultrastructure
Collapse
Affiliation(s)
- Roos Anouk Voorn
- Presynaptogenesis and Intracellular Transport in Hair Cells Junior Research Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Goettingen, 37075 Goettingen, Germany;
- Göttingen Graduate Center for Neurosciences, Biophysics and Molecular Biosciences, 37075 Goettingen, Germany
- Collaborative Research Center 889 “Cellular Mechanisms of Sensory Processing”, 37075 Goettingen, Germany
| | - Christian Vogl
- Presynaptogenesis and Intracellular Transport in Hair Cells Junior Research Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Goettingen, 37075 Goettingen, Germany;
- Collaborative Research Center 889 “Cellular Mechanisms of Sensory Processing”, 37075 Goettingen, Germany
| |
Collapse
|
25
|
Dembla E, Dembla M, Maxeiner S, Schmitz F. Synaptic ribbons foster active zone stability and illumination-dependent active zone enrichment of RIM2 and Cav1.4 in photoreceptor synapses. Sci Rep 2020; 10:5957. [PMID: 32249787 PMCID: PMC7136232 DOI: 10.1038/s41598-020-62734-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 03/18/2020] [Indexed: 02/08/2023] Open
Abstract
Rod photoreceptor synapses use large, ribbon-type active zones for continuous synaptic transmission during light and dark. Since ribbons are physically connected to the active zones, we asked whether illumination-dependent changes of ribbons influence Cav1.4/RIM2 protein clusters at the active zone and whether these illumination-dependent effects at the active zone require the presence of the synaptic ribbon. We found that synaptic ribbon length and the length of presynaptic Cav1.4/RIM2 clusters are tightly correlated. Dark-adaptation did not change the number of ribbons and active zone puncta. However, mean ribbon length and length of presynaptic Cav1.4/RIM2 clusters increased significantly during dark-adaptation when tonic exocytosis is highest. In the present study, we identified by the analyses of synaptic ribbon-deficient RIBEYE knockout mice that synaptic ribbons are (1) needed to stabilize Cav1.4/RIM2 at rod photoreceptor active zones and (2) are required for the darkness-induced active zone enrichment of Cav1.4/RIM2. These data propose a role of the ribbon in active zone stabilization and suggest a homeostatic function of the ribbon in illumination-dependent active zone remodeling.
Collapse
Affiliation(s)
- Ekta Dembla
- Institute of Anatomy and Cell Biology, Department of Neuroanatomy, Saarland University, Medical School, 66421, Homburg, Germany.
| | - Mayur Dembla
- Institute of Anatomy and Cell Biology, Department of Neuroanatomy, Saarland University, Medical School, 66421, Homburg, Germany
| | - Stephan Maxeiner
- Institute of Anatomy and Cell Biology, Department of Neuroanatomy, Saarland University, Medical School, 66421, Homburg, Germany
- Institute of Anatomy and Cell Biology, Saarland University, AG Krasteva-Christ, 66421, Homburg, Germany
| | - Frank Schmitz
- Institute of Anatomy and Cell Biology, Department of Neuroanatomy, Saarland University, Medical School, 66421, Homburg, Germany.
| |
Collapse
|
26
|
Erickson T, Pacentine IV, Venuto A, Clemens R, Nicolson T. The lhfpl5 Ohnologs lhfpl5a and lhfpl5b Are Required for Mechanotransduction in Distinct Populations of Sensory Hair Cells in Zebrafish. Front Mol Neurosci 2020; 12:320. [PMID: 32009898 PMCID: PMC6974483 DOI: 10.3389/fnmol.2019.00320] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 12/16/2019] [Indexed: 01/20/2023] Open
Abstract
Hair cells sense and transmit auditory, vestibular, and hydrodynamic information by converting mechanical stimuli into electrical signals. This process of mechano-electrical transduction (MET) requires a mechanically gated channel localized in the apical stereocilia of hair cells. In mice, lipoma HMGIC fusion partner-like 5 (LHFPL5) acts as an auxiliary subunit of the MET channel whose primary role is to correctly localize PCDH15 and TMC1 to the mechanotransduction complex. Zebrafish have two lhfpl5 genes (lhfpl5a and lhfpl5b), but their individual contributions to MET channel assembly and function have not been analyzed. Here we show that the zebrafish lhfpl5 genes are expressed in discrete populations of hair cells: lhfpl5a expression is restricted to auditory and vestibular hair cells in the inner ear, while lhfpl5b expression is specific to hair cells of the lateral line organ. Consequently, lhfpl5a mutants exhibit defects in auditory and vestibular function, while disruption of lhfpl5b affects hair cells only in the lateral line neuromasts. In contrast to previous reports in mice, localization of Tmc1 does not depend upon Lhfpl5 function in either the inner ear or lateral line organ. In both lhfpl5a and lhfpl5b mutants, GFP-tagged Tmc1 and Tmc2b proteins still localize to the stereocilia of hair cells. Using a stably integrated GFP-Lhfpl5a transgene, we show that the tip link cadherins Pcdh15a and Cdh23, along with the Myo7aa motor protein, are required for correct Lhfpl5a localization at the tips of stereocilia. Our work corroborates the evolutionarily conserved co-dependence between Lhfpl5 and Pcdh15, but also reveals novel requirements for Cdh23 and Myo7aa to correctly localize Lhfpl5a. In addition, our data suggest that targeting of Tmc1 and Tmc2b proteins to stereocilia in zebrafish hair cells occurs independently of Lhfpl5 proteins.
Collapse
Affiliation(s)
- Timothy Erickson
- Department of Biology, East Carolina University, Greenville, NC, United States.,Oregon Hearing Research Center and Vollum Institute, Oregon Health and Science University, Portland, OR, United States
| | - Itallia V Pacentine
- Oregon Hearing Research Center and Vollum Institute, Oregon Health and Science University, Portland, OR, United States
| | - Alexandra Venuto
- Department of Biology, East Carolina University, Greenville, NC, United States
| | - Rachel Clemens
- Oregon Hearing Research Center and Vollum Institute, Oregon Health and Science University, Portland, OR, United States
| | - Teresa Nicolson
- Oregon Hearing Research Center and Vollum Institute, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
27
|
Moser T, Grabner CP, Schmitz F. Sensory Processing at Ribbon Synapses in the Retina and the Cochlea. Physiol Rev 2020; 100:103-144. [DOI: 10.1152/physrev.00026.2018] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In recent years, sensory neuroscientists have made major efforts to dissect the structure and function of ribbon synapses which process sensory information in the eye and ear. This review aims to summarize our current understanding of two key aspects of ribbon synapses: 1) their mechanisms of exocytosis and endocytosis and 2) their molecular anatomy and physiology. Our comparison of ribbon synapses in the cochlea and the retina reveals convergent signaling mechanisms, as well as divergent strategies in different sensory systems.
Collapse
Affiliation(s)
- Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany; Auditory Neuroscience Group, Max Planck Institute for Experimental Medicine, Göttingen, Germany; Synaptic Nanophysiology Group, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany; and Institute for Anatomy and Cell Biology, Department of Neuroanatomy, Medical School, Saarland University, Homburg, Germany
| | - Chad P. Grabner
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany; Auditory Neuroscience Group, Max Planck Institute for Experimental Medicine, Göttingen, Germany; Synaptic Nanophysiology Group, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany; and Institute for Anatomy and Cell Biology, Department of Neuroanatomy, Medical School, Saarland University, Homburg, Germany
| | - Frank Schmitz
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany; Auditory Neuroscience Group, Max Planck Institute for Experimental Medicine, Göttingen, Germany; Synaptic Nanophysiology Group, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany; and Institute for Anatomy and Cell Biology, Department of Neuroanatomy, Medical School, Saarland University, Homburg, Germany
| |
Collapse
|
28
|
Wong HTC, Zhang Q, Beirl AJ, Petralia RS, Wang YX, Kindt K. Synaptic mitochondria regulate hair-cell synapse size and function. eLife 2019; 8:e48914. [PMID: 31609202 PMCID: PMC6879205 DOI: 10.7554/elife.48914] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 10/13/2019] [Indexed: 11/21/2022] Open
Abstract
Sensory hair cells in the ear utilize specialized ribbon synapses. These synapses are defined by electron-dense presynaptic structures called ribbons, composed primarily of the structural protein Ribeye. Previous work has shown that voltage-gated influx of Ca2+ through CaV1.3 channels is critical for hair-cell synapse function and can impede ribbon formation. We show that in mature zebrafish hair cells, evoked presynaptic-Ca2+ influx through CaV1.3 channels initiates mitochondrial-Ca2+ (mito-Ca2+) uptake adjacent to ribbons. Block of mito-Ca2+ uptake in mature cells depresses presynaptic-Ca2+ influx and impacts synapse integrity. In developing zebrafish hair cells, mito-Ca2+ uptake coincides with spontaneous rises in presynaptic-Ca2+ influx. Spontaneous mito-Ca2+ loading lowers cellular NAD+/NADH redox and downregulates ribbon size. Direct application of NAD+ or NADH increases or decreases ribbon size respectively, possibly acting through the NAD(H)-binding domain on Ribeye. Our results present a mechanism where presynaptic- and mito-Ca2+ couple to confer proper presynaptic function and formation.
Collapse
MESH Headings
- 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology
- Animals
- Animals, Genetically Modified
- Calcium/metabolism
- Calcium Channel Agonists/pharmacology
- Calcium Channel Blockers/pharmacology
- Calcium Channels, L-Type/genetics
- Calcium Channels, L-Type/metabolism
- Calcium Signaling
- Cell Size
- Embryo, Nonmammalian
- Evoked Potentials, Auditory/physiology
- Eye Proteins/chemistry
- Eye Proteins/genetics
- Eye Proteins/metabolism
- Gene Expression
- Hair Cells, Auditory/cytology
- Hair Cells, Auditory/drug effects
- Hair Cells, Auditory/metabolism
- Isradipine/pharmacology
- Mitochondria/drug effects
- Mitochondria/metabolism
- Mitochondria/ultrastructure
- NAD/metabolism
- Oxidation-Reduction
- Protein Binding
- Protein Interaction Domains and Motifs
- Ruthenium Compounds/pharmacology
- Synapses/drug effects
- Synapses/metabolism
- Synapses/ultrastructure
- Synaptic Transmission
- Zebrafish
- Zebrafish Proteins/agonists
- Zebrafish Proteins/antagonists & inhibitors
- Zebrafish Proteins/chemistry
- Zebrafish Proteins/genetics
- Zebrafish Proteins/metabolism
Collapse
Affiliation(s)
- Hiu-tung C Wong
- Section on Sensory Cell Development and FunctionNational Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
- National Institutes of Health-Johns Hopkins University Graduate Partnership ProgramNational Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | - Qiuxiang Zhang
- Section on Sensory Cell Development and FunctionNational Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | - Alisha J Beirl
- Section on Sensory Cell Development and FunctionNational Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | - Ronald S Petralia
- Advanced Imaging CoreNational Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | - Ya-Xian Wang
- Advanced Imaging CoreNational Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | - Katie Kindt
- Section on Sensory Cell Development and FunctionNational Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| |
Collapse
|
29
|
Manchanda A, Chatterjee P, Bonventre JA, Haggard DE, Kindt KS, Tanguay RL, Johnson CP. Otoferlin Depletion Results in Abnormal Synaptic Ribbons and Altered Intracellular Calcium Levels in Zebrafish. Sci Rep 2019; 9:14273. [PMID: 31582816 PMCID: PMC6776657 DOI: 10.1038/s41598-019-50710-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 09/13/2019] [Indexed: 01/10/2023] Open
Abstract
The protein otoferlin plays an essential role at the sensory hair cell synapse. Mutations in otoferlin result in deafness and depending on the species, mild to strong vestibular deficits. While studies in mouse models suggest a role for otoferlin in synaptic vesicle exocytosis and endocytosis, it is unclear whether these functions are conserved across species. To address this question, we characterized the impact of otoferlin depletion in zebrafish larvae and found defects in synaptic vesicle recycling, abnormal synaptic ribbons, and higher resting calcium concentrations in hair cells. We also observed abnormal expression of the calcium binding hair cell genes s100s and parvalbumin, as well as the nogo related proteins rtn4rl2a and rtn4rl2b. Exogenous otoferlin partially restored expression of genes affected by endogenous otoferlin depletion. Our results suggest that in addition to vesicle recycling, depletion of otoferlin disrupts resting calcium levels, alters synaptic ribbon architecture, and perturbs transcription of hair cells specific genes during zebrafish development.
Collapse
Affiliation(s)
- Aayushi Manchanda
- Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon, USA
| | - Paroma Chatterjee
- Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon, USA
| | - Josephine A Bonventre
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon, USA
| | - Derik E Haggard
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon, USA
| | - Katie S Kindt
- National Institute of Deafness and Other Communication Disorders (NIDCD), NIH, Maryland, USA
| | - Robert L Tanguay
- Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon, USA
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon, USA
| | - Colin P Johnson
- Molecular and Cellular Biology Program, Oregon State University, Corvallis, Oregon, USA.
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon, USA.
| |
Collapse
|
30
|
Wells HRR, Freidin MB, Zainul Abidin FN, Payton A, Dawes P, Munro KJ, Morton CC, Moore DR, Dawson SJ, Williams FMK. GWAS Identifies 44 Independent Associated Genomic Loci for Self-Reported Adult Hearing Difficulty in UK Biobank. Am J Hum Genet 2019; 105:788-802. [PMID: 31564434 PMCID: PMC6817556 DOI: 10.1016/j.ajhg.2019.09.008] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 09/04/2019] [Indexed: 01/13/2023] Open
Abstract
Age-related hearing impairment (ARHI) is the most common sensory impairment in the aging population; a third of individuals are affected by disabling hearing loss by the age of 65. It causes social isolation and depression and has recently been identified as a risk factor for dementia. The genetic risk factors and underlying pathology of ARHI are largely unknown, meaning that targets for new therapies remain elusive, yet heritability estimates range between 35% and 55%. We performed genome-wide association studies (GWASs) for two self-reported hearing phenotypes, using more than 250,000 UK Biobank (UKBB) volunteers aged between 40 and 69 years. Forty-four independent genome-wide significant loci (p < 5E-08) were identified, considerably increasing the number of established trait loci. Thirty-four loci are novel associations with hearing loss of any form, and only one of the ten known hearing loci has a previously reported association with an ARHI-related trait. Gene sets from these loci are enriched in auditory processes such as synaptic activities, nervous system processes, inner ear morphology, and cognition, while genetic correlation analysis revealed strong positive correlations with multiple personality and psychological traits for the first time. Immunohistochemistry for protein localization in adult mouse cochlea implicate metabolic, sensory, and neuronal functions for NID2, CLRN2, and ARHGEF28. These results provide insight into the genetic landscape underlying ARHI, opening up novel therapeutic targets for further investigation. In a wider context, our study also highlights the viability of using self-report phenotypes for genetic discovery in very large samples when deep phenotyping is unavailable.
Collapse
Affiliation(s)
- Helena R R Wells
- Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King's College London, London SE1 7EH, UK; UCL Ear Institute, University College London, London WC1X 8EE, UK
| | - Maxim B Freidin
- Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King's College London, London SE1 7EH, UK
| | - Fatin N Zainul Abidin
- UCL Ear Institute, University College London, London WC1X 8EE, UK; Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, WC1E 7JE, UK
| | - Antony Payton
- Division of Informatics, Imaging & Data Sciences, The University of Manchester, Manchester M13 9PT, UK
| | - Piers Dawes
- Manchester Centre for Audiology and Deafness, The University of Manchester, Manchester M13 9PL, UK
| | - Kevin J Munro
- Manchester Centre for Audiology and Deafness, The University of Manchester, Manchester M13 9PL, UK; Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9WL, UK
| | - Cynthia C Morton
- Manchester Centre for Audiology and Deafness, The University of Manchester, Manchester M13 9PL, UK; Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9WL, UK; Departments of Obstetrics and Gynecology and of Pathology, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02115, USA
| | - David R Moore
- Manchester Centre for Audiology and Deafness, The University of Manchester, Manchester M13 9PL, UK; Cincinnati Children's Hospital Medical Centre, Department of Otolaryngology, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Sally J Dawson
- UCL Ear Institute, University College London, London WC1X 8EE, UK.
| | - Frances M K Williams
- Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King's College London, London SE1 7EH, UK.
| |
Collapse
|
31
|
Okawa H, Yu WQ, Matti U, Schwarz K, Odermatt B, Zhong H, Tsukamoto Y, Lagnado L, Rieke F, Schmitz F, Wong ROL. Dynamic assembly of ribbon synapses and circuit maintenance in a vertebrate sensory system. Nat Commun 2019; 10:2167. [PMID: 31092821 PMCID: PMC6520400 DOI: 10.1038/s41467-019-10123-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 04/10/2019] [Indexed: 11/11/2022] Open
Abstract
Ribbon synapses transmit information in sensory systems, but their development is not well understood. To test the hypothesis that ribbon assembly stabilizes nascent synapses, we performed simultaneous time-lapse imaging of fluorescently-tagged ribbons in retinal cone bipolar cells (BCs) and postsynaptic densities (PSD95-FP) of retinal ganglion cells (RGCs). Ribbons and PSD95-FP clusters were more stable when these components colocalized at synapses. However, synapse density on ON-alpha RGCs was unchanged in mice lacking ribbons (ribeye knockout). Wildtype BCs make both ribbon-containing and ribbon-free synapses with these GCs even at maturity. Ribbon assembly and cone BC-RGC synapse maintenance are thus regulated independently. Despite the absence of synaptic ribbons, RGCs continued to respond robustly to light stimuli, although quantitative examination of the responses revealed reduced frequency and contrast sensitivity.
Collapse
Affiliation(s)
- Haruhisa Okawa
- Department of Biological Structure, University of Washington, Seattle, 98195, WA, USA
| | - Wan-Qing Yu
- Department of Biological Structure, University of Washington, Seattle, 98195, WA, USA
| | - Ulf Matti
- Department of Neuroanatomy, Medical School Homburg/Saar, Institute for Anatomy and Cell Biology, Saarland University, Homburg/Saar, 66421, Germany
| | - Karin Schwarz
- Department of Neuroanatomy, Medical School Homburg/Saar, Institute for Anatomy and Cell Biology, Saarland University, Homburg/Saar, 66421, Germany
| | | | - Haining Zhong
- Vollum institute, Oregon Health and Science University, Portland, 97239, OR, USA
| | - Yoshihiko Tsukamoto
- Department of Biology, Hyogo College of Medicine, Nishinomiya, 663-8501, Hyogo, Japan
| | - Leon Lagnado
- School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - Fred Rieke
- Department of Physiology and Biophysics, University of Washington, Seattle, 98195, WA, USA
| | - Frank Schmitz
- Department of Neuroanatomy, Medical School Homburg/Saar, Institute for Anatomy and Cell Biology, Saarland University, Homburg/Saar, 66421, Germany
| | - Rachel O L Wong
- Department of Biological Structure, University of Washington, Seattle, 98195, WA, USA.
| |
Collapse
|
32
|
Coate TM, Scott MK, Gurjar MC. Current concepts in cochlear ribbon synapse formation. Synapse 2019; 73:e22087. [PMID: 30592086 PMCID: PMC6573016 DOI: 10.1002/syn.22087] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 12/20/2018] [Accepted: 12/21/2018] [Indexed: 12/11/2022]
Abstract
In mammals, hair cells and spiral ganglion neurons (SGNs) in the cochlea together are sophisticated "sensorineural" structures that transduce auditory information from the outside world into the brain. Hair cells and SGNs are joined by glutamatergic ribbon-type synapses composed of a molecular machinery rivaling in complexity the mechanoelectric transduction components found at the apical side of the hair cell. The cochlear hair cell ribbon synapse has received much attention lately because of recent and important findings related to its damage (sometimes termed "synaptopathy") as a result of noise overexposure. During development, ribbon synapses between type I SGNs and inner hair cells form in the time window between birth and hearing onset and is a process coordinated with type I SGN myelination, spontaneous activity, synaptic pruning, and innervation by efferents. In this review, we highlight new findings regarding the diversity of type I SGNs and inner hair cell synapses, and the molecular mechanisms of selective hair cell targeting. Also discussed are cell adhesion molecules and protein constituents of the ribbon synapse, and how these factors participate in ribbon synapse formation. We also note interesting new insights into the morphological development of type II SGNs, and the potential for cochlear macrophages as important players in protecting SGNs. We also address recent studies demonstrating that the structural and physiological profiles of the type I SGNs do not reach full maturity until weeks after hearing onset, suggesting a protracted development that is likely modulated by activity.
Collapse
Affiliation(s)
- Thomas M. Coate
- Georgetown University, Department of Biology, 37th and O St. NW. Washington, DC. 20007. USA
| | - M. Katie Scott
- Department of Biological Sciences and Purdue Institute of Integrative Neuroscience, Purdue University, West Lafayette, Indiana 47907. USA
| | - Mansa C. Gurjar
- Georgetown University, Department of Biology, 37th and O St. NW. Washington, DC. 20007. USA
| |
Collapse
|
33
|
Chakrabarti R, Wichmann C. Nanomachinery Organizing Release at Neuronal and Ribbon Synapses. Int J Mol Sci 2019; 20:E2147. [PMID: 31052288 PMCID: PMC6539712 DOI: 10.3390/ijms20092147] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 04/26/2019] [Accepted: 04/26/2019] [Indexed: 11/17/2022] Open
Abstract
A critical aim in neuroscience is to obtain a comprehensive view of how regulated neurotransmission is achieved. Our current understanding of synapses relies mainly on data from electrophysiological recordings, imaging, and molecular biology. Based on these methodologies, proteins involved in a synaptic vesicle (SV) formation, mobility, and fusion at the active zone (AZ) membrane have been identified. In the last decade, electron tomography (ET) combined with a rapid freezing immobilization of neuronal samples opened a window for understanding the structural machinery with the highest spatial resolution in situ. ET provides significant insights into the molecular architecture of the AZ and the organelles within the presynaptic nerve terminal. The specialized sensory ribbon synapses exhibit a distinct architecture from neuronal synapses due to the presence of the electron-dense synaptic ribbon. However, both synapse types share the filamentous structures, also commonly termed as tethers that are proposed to contribute to different steps of SV recruitment and exocytosis. In this review, we discuss the emerging views on the role of filamentous structures in SV exocytosis gained from ultrastructural studies of excitatory, mainly central neuronal compared to ribbon-type synapses with a focus on inner hair cell (IHC) ribbon synapses. Moreover, we will speculate on the molecular entities that may be involved in filament formation and hence play a crucial role in the SV cycle.
Collapse
Affiliation(s)
- Rituparna Chakrabarti
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany.
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany.
- Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing", 37099 Göttingen, Germany.
| | - Carolin Wichmann
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany.
- Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany.
- Collaborative Research Center 889 "Cellular Mechanisms of Sensory Processing", 37099 Göttingen, Germany.
- Collaborative Research Center 1286 "Quantitative Synaptology", 37099 Göttingen, Germany.
- Auditory Neuroscience Group, Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany.
| |
Collapse
|
34
|
Intrinsic planar polarity mechanisms influence the position-dependent regulation of synapse properties in inner hair cells. Proc Natl Acad Sci U S A 2019; 116:9084-9093. [PMID: 30975754 DOI: 10.1073/pnas.1818358116] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Encoding the wide range of audible sounds in the mammalian cochlea is collectively achieved by functionally diverse type I spiral ganglion neurons (SGNs) at each tonotopic position. The firing of each SGN is thought to be driven by an individual active zone (AZ) of a given inner hair cell (IHC). These AZs present distinct properties according to their position within the IHC, to some extent forming a gradient between the modiolar and the pillar IHC side. In this study, we investigated whether signaling involved in planar polarity at the apical surface can influence position-dependent AZ properties at the IHC base. Specifically, we tested the role of Gαi proteins and their binding partner LGN/Gpsm2 implicated in cytoskeleton polarization and hair cell (HC) orientation along the epithelial plane. Using high and superresolution immunofluorescence microscopy as well as patch-clamp combined with confocal Ca2+ imaging we analyzed IHCs in which Gαi signaling was blocked by Cre-induced expression of the pertussis toxin catalytic subunit (PTXa). PTXa-expressing IHCs exhibited larger CaV1.3 Ca2+-channel clusters and consequently greater Ca2+ influx at the whole-cell and single-synapse levels, which also showed a hyperpolarized shift of activation. Moreover, PTXa expression collapsed the modiolar-pillar gradients of ribbon size and maximal synaptic Ca2+ influx. Finally, genetic deletion of Gαi3 and LGN/Gpsm2 also disrupted the modiolar-pillar gradient of ribbon size. We propose a role for Gαi proteins and LGN in regulating the position-dependent AZ properties in IHCs and suggest that this signaling pathway contributes to setting up the diverse firing properties of SGNs.
Collapse
|
35
|
Michanski S, Smaluch K, Steyer AM, Chakrabarti R, Setz C, Oestreicher D, Fischer C, Möbius W, Moser T, Vogl C, Wichmann C. Mapping developmental maturation of inner hair cell ribbon synapses in the apical mouse cochlea. Proc Natl Acad Sci U S A 2019; 116:6415-6424. [PMID: 30867284 PMCID: PMC6442603 DOI: 10.1073/pnas.1812029116] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Ribbon synapses of cochlear inner hair cells (IHCs) undergo molecular assembly and extensive functional and structural maturation before hearing onset. Here, we characterized the nanostructure of IHC synapses from late prenatal mouse embryo stages (embryonic days 14-18) into adulthood [postnatal day (P)48] using electron microscopy and tomography as well as optical nanoscopy of apical turn organs of Corti. We find that synaptic ribbon precursors arrive at presynaptic active zones (AZs) after afferent contacts have been established. These ribbon precursors contain the proteins RIBEYE and piccolino, tether synaptic vesicles and their delivery likely involves active, microtubule-based transport pathways. Synaptic contacts undergo a maturational transformation from multiple small to one single, large AZ. This maturation is characterized by the fusion of ribbon precursors with membrane-anchored ribbons that also appear to fuse with each other. Such fusion events are most frequently encountered around P12 and hence, coincide with hearing onset in mice. Thus, these events likely underlie the morphological and functional maturation of the AZ. Moreover, the postsynaptic densities appear to undergo a similar refinement alongside presynaptic maturation. Blockwise addition of ribbon material by fusion as found during AZ maturation might represent a general mechanism for modulating ribbon size.
Collapse
Affiliation(s)
- Susann Michanski
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience, InnerEarLab and Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, 37075 Göttingen, Germany
| | - Katharina Smaluch
- Collaborative Research Center 889, University of Göttingen, 37075 Göttingen, Germany
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany
- Presynaptogenesis and Intracellular Transport in Hair Cells Junior Research Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Anna Maria Steyer
- Electron Microscopy Core Unit, Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
- Center Nanoscale Microscopy and Molecular Physiology of the Brain, University of Göttingen, 37075 Göttingen, Germany
| | - Rituparna Chakrabarti
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience, InnerEarLab and Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, 37075 Göttingen, Germany
| | - Cristian Setz
- Collaborative Research Center 889, University of Göttingen, 37075 Göttingen, Germany
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany
- Presynaptogenesis and Intracellular Transport in Hair Cells Junior Research Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany
- Department of Otolaryngology, Head and Neck Surgery, University Medical Center Göttingen, 37075 Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - David Oestreicher
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany
- Presynaptogenesis and Intracellular Transport in Hair Cells Junior Research Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany
- Department of Otolaryngology, Head and Neck Surgery, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Christian Fischer
- Johann Friedrich Blumenbach Institute for Zoology and Anthropology, Department of Animal Evolution and Biodiversity, Georg August University of Göttingen, 37073 Göttingen, Germany
| | - Wiebke Möbius
- Electron Microscopy Core Unit, Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
- Center Nanoscale Microscopy and Molecular Physiology of the Brain, University of Göttingen, 37075 Göttingen, Germany
| | - Tobias Moser
- Collaborative Research Center 889, University of Göttingen, 37075 Göttingen, Germany
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany
- Center Nanoscale Microscopy and Molecular Physiology of the Brain, University of Göttingen, 37075 Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
- Synaptic Nanophysiology Group, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
| | - Christian Vogl
- Collaborative Research Center 889, University of Göttingen, 37075 Göttingen, Germany;
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany
- Presynaptogenesis and Intracellular Transport in Hair Cells Junior Research Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Carolin Wichmann
- Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience, InnerEarLab and Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37075 Göttingen, Germany;
- Collaborative Research Center 889, University of Göttingen, 37075 Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| |
Collapse
|
36
|
Pickett SB, Raible DW. Water Waves to Sound Waves: Using Zebrafish to Explore Hair Cell Biology. J Assoc Res Otolaryngol 2019; 20:1-19. [PMID: 30635804 DOI: 10.1007/s10162-018-00711-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 12/19/2018] [Indexed: 01/09/2023] Open
Abstract
Although perhaps best known for their use in developmental studies, over the last couple of decades, zebrafish have become increasingly popular model organisms for investigating auditory system function and disease. Like mammals, zebrafish possess inner ear mechanosensory hair cells required for hearing, as well as superficial hair cells of the lateral line sensory system, which mediate detection of directional water flow. Complementing mammalian studies, zebrafish have been used to gain significant insights into many facets of hair cell biology, including mechanotransduction and synaptic physiology as well as mechanisms of both hereditary and acquired hair cell dysfunction. Here, we provide an overview of this literature, highlighting some of the particular advantages of using zebrafish to investigate hearing and hearing loss.
Collapse
Affiliation(s)
- Sarah B Pickett
- Department of Biological Structure, University of Washington, Health Sciences Building H-501, 1959 NE Pacific Street, Box 357420, Seattle, WA, 98195-7420, USA
- Graduate Program in Neuroscience, University of Washington, 1959 NE Pacific Street, Box 357270, Seattle, WA, 98195-7270, USA
| | - David W Raible
- Department of Biological Structure, University of Washington, Health Sciences Building H-501, 1959 NE Pacific Street, Box 357420, Seattle, WA, 98195-7420, USA.
- Graduate Program in Neuroscience, University of Washington, 1959 NE Pacific Street, Box 357270, Seattle, WA, 98195-7270, USA.
- Virginia Merrill Bloedel Hearing Research Center, University of Washington, 1701 NE Columbia Rd, Box 357923, Seattle, WA, 98195-7923, USA.
| |
Collapse
|
37
|
Pangrsic T, Singer JH, Koschak A. Voltage-Gated Calcium Channels: Key Players in Sensory Coding in the Retina and the Inner Ear. Physiol Rev 2019; 98:2063-2096. [PMID: 30067155 DOI: 10.1152/physrev.00030.2017] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Calcium influx through voltage-gated Ca (CaV) channels is the first step in synaptic transmission. This review concerns CaV channels at ribbon synapses in primary sense organs and their specialization for efficient coding of stimuli in the physical environment. Specifically, we describe molecular, biochemical, and biophysical properties of the CaV channels in sensory receptor cells of the retina, cochlea, and vestibular apparatus, and we consider how such properties might change over the course of development and contribute to synaptic plasticity. We pay particular attention to factors affecting the spatial arrangement of CaV channels at presynaptic, ribbon-type active zones, because the spatial relationship between CaV channels and release sites has been shown to affect synapse function critically in a number of systems. Finally, we review identified synaptopathies affecting sensory systems and arising from dysfunction of L-type, CaV1.3, and CaV1.4 channels or their protein modulatory elements.
Collapse
Affiliation(s)
- Tina Pangrsic
- Synaptic Physiology of Mammalian Vestibular Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen and Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine , Göttingen, Germany ; Department of Biology, University of Maryland , College Park, Maryland ; and Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck , Innsbruck , Austria
| | - Joshua H Singer
- Synaptic Physiology of Mammalian Vestibular Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen and Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine , Göttingen, Germany ; Department of Biology, University of Maryland , College Park, Maryland ; and Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck , Innsbruck , Austria
| | - Alexandra Koschak
- Synaptic Physiology of Mammalian Vestibular Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen and Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine , Göttingen, Germany ; Department of Biology, University of Maryland , College Park, Maryland ; and Pharmacology and Toxicology, Institute of Pharmacy, University of Innsbruck , Innsbruck , Austria
| |
Collapse
|
38
|
Yu Y, Hu B, Bao J, Mulvany J, Bielefeld E, Harrison RT, Neton SA, Thirumala P, Chen Y, Lei D, Qiu Z, Zheng Q, Ren J, Perez-Flores MC, Yamoah EN, Salehi P. Otoprotective Effects of Stephania tetrandra S. Moore Herb Isolate against Acoustic Trauma. J Assoc Res Otolaryngol 2018; 19:653-668. [PMID: 30187298 PMCID: PMC6249158 DOI: 10.1007/s10162-018-00690-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 07/08/2018] [Indexed: 01/10/2023] Open
Abstract
Noise is the most common occupational and environmental hazard, and noise-induced hearing loss (NIHL) is the second most common form of sensorineural hearing deficit. Although therapeutics that target the free-radical pathway have shown promise, none of these compounds is currently approved against NIHL by the United States Food and Drug Administration. The present study has demonstrated that tetrandrine (TET), a traditional Chinese medicinal alkaloid and the main chemical isolate of the Stephania tetrandra S. Moore herb, significantly attenuated NIHL in CBA/CaJ mice. TET is known to exert antihypertensive and antiarrhythmic effects through the blocking of calcium channels. Whole-cell patch-clamp recording from adult spiral ganglion neurons showed that TET blocked the transient Ca2+ current in a dose-dependent manner and the half-blocking concentration was 0.6 + 0.1 μM. Consistent with previous findings that modulations of calcium-based signaling pathways have both prophylactic and therapeutic effects against neural trauma, NIHL was significantly diminished by TET administration. Importantly, TET has a long-lasting protective effect after noise exposure (48 weeks) in comparison to 2 weeks after noise exposure. The otoprotective effects of TET were achieved mainly by preventing outer hair cell damage and synapse loss between inner hair cells and spiral ganglion neurons. Thus, our data indicate that TET has great potential in the prevention and treatment of NIHL.
Collapse
Affiliation(s)
- Yan Yu
- The First People’s Hospital of Zhangjiagang, 68 W Jiyang Road, Zhangjiagang City, 215600 Jiangsu China
- Translational Research Center, Northeast Ohio Medical University, Rootstown, OH 44272 USA
| | - Bing Hu
- Translational Research Center, Northeast Ohio Medical University, Rootstown, OH 44272 USA
- Department of Otolaryngology-Head and Neck Surgery, Case Western Reserve University School of Medicine, Cleveland, OH 44106 USA
- Department of Otolaryngology Head and Neck Surgery, The Second Xiangya Hospital of Central South University, Changsha, 440011 Hunan China
| | - Jianxin Bao
- Translational Research Center, Northeast Ohio Medical University, Rootstown, OH 44272 USA
- Department of Research and Development, Gateway Biotechnology Inc., Rootstown, OH 44272 USA
| | - Jessica Mulvany
- Translational Research Center, Northeast Ohio Medical University, Rootstown, OH 44272 USA
- Department of Research and Development, Gateway Biotechnology Inc., Rootstown, OH 44272 USA
| | - Eric Bielefeld
- Department of Speech and Hearing Science, Ohio State University, Columbus, OH 43210 USA
| | - Ryan T. Harrison
- Department of Speech and Hearing Science, Ohio State University, Columbus, OH 43210 USA
| | - Sarah A. Neton
- Department of Speech and Hearing Science, Ohio State University, Columbus, OH 43210 USA
| | - Partha Thirumala
- The University of Pittsburgh Medical Center, Suite B-400, 200 Lothrop Street, Pittsburgh, PA 15213 USA
| | - Yingying Chen
- Translational Research Center, Northeast Ohio Medical University, Rootstown, OH 44272 USA
| | - Debin Lei
- Translational Research Center, Northeast Ohio Medical University, Rootstown, OH 44272 USA
| | - Ziyu Qiu
- Department of Research and Development, Gateway Biotechnology Inc., Rootstown, OH 44272 USA
| | - Qingyin Zheng
- Department of Otolaryngology-Head and Neck Surgery, Case Western Reserve University School of Medicine, Cleveland, OH 44106 USA
| | - Jihao Ren
- Department of Otolaryngology Head and Neck Surgery, The Second Xiangya Hospital of Central South University, Changsha, 440011 Hunan China
| | - Maria Cristina Perez-Flores
- Department of Physiology and Cell Biology, University of Nevada Reno, 1664 North Virginia St, Reno, NV 89557 USA
| | - Ebenezer N. Yamoah
- Department of Physiology and Cell Biology, University of Nevada Reno, 1664 North Virginia St, Reno, NV 89557 USA
| | - Pezhman Salehi
- Translational Research Center, Northeast Ohio Medical University, Rootstown, OH 44272 USA
| |
Collapse
|
39
|
Pangrsic T, Vogl C. Balancing presynaptic release and endocytic membrane retrieval at hair cell ribbon synapses. FEBS Lett 2018; 592:3633-3650. [PMID: 30251250 DOI: 10.1002/1873-3468.13258] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/18/2018] [Accepted: 09/19/2018] [Indexed: 11/07/2022]
Abstract
The timely and reliable processing of auditory and vestibular information within the inner ear requires highly sophisticated sensory transduction pathways. On a cellular level, these demands are met by hair cells, which respond to sound waves - or alterations in body positioning - by releasing glutamate-filled synaptic vesicles (SVs) from their presynaptic active zones with unprecedented speed and exquisite temporal fidelity, thereby initiating the auditory and vestibular pathways. In order to achieve this, hair cells have developed anatomical and molecular specializations, such as the characteristic and name-giving 'synaptic ribbons' - presynaptically anchored dense bodies that tether SVs prior to release - as well as other unique or unconventional synaptic proteins. The tightly orchestrated interplay between these molecular components enables not only ultrafast exocytosis, but similarly rapid and efficient compensatory endocytosis. So far, the knowledge of how endocytosis operates at hair cell ribbon synapses is limited. In this Review, we summarize recent advances in our understanding of the SV cycle and molecular anatomy of hair cell ribbon synapses, with a focus on cochlear inner hair cells.
Collapse
Affiliation(s)
- Tina Pangrsic
- Synaptic Physiology of Mammalian Vestibular Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine, University Medical Center Göttingen, Germany
| | - Christian Vogl
- Presynaptogenesis and Intracellular Transport in Hair Cells Group, Institute for Auditory Neuroscience and InnerEarLab, Auditory Neuroscience Group, Max Planck Institute of Experimental Medicine, University Medical Center Göttingen, Germany
| |
Collapse
|
40
|
Kindt KS, Sheets L. Transmission Disrupted: Modeling Auditory Synaptopathy in Zebrafish. Front Cell Dev Biol 2018; 6:114. [PMID: 30258843 PMCID: PMC6143809 DOI: 10.3389/fcell.2018.00114] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/23/2018] [Indexed: 01/04/2023] Open
Abstract
Sensorineural hearing loss is the most common form of hearing loss in humans, and results from either dysfunction in hair cells, the sensory receptors of sound, or the neurons that innervate hair cells. A specific type of sensorineural hearing loss, referred to as auditory synaptopathy, occurs when hair cells are able to detect sound but fail to transmit sound stimuli at the hair-cell synapse. Auditory synaptopathy can originate from genetic alterations that specifically disrupt hair-cell synapse function. Additionally, environmental factors such as noise exposure can leave hair cells intact but result in loss of hair-cell synapses, and represent an acquired form of auditory synaptopathy. The zebrafish model has emerged as a valuable system for studies of hair-cell function, and specifically hair-cell synaptopathy. In this review, we describe the experimental tools that have been developed to study hair-cell synapses in zebrafish. We discuss how zebrafish genetics has helped identify and define the roles of hair-cell synaptic proteins crucial for hearing in humans, and highlight how studies in zebrafish have contributed to our understanding of hair-cell synapse formation and function. In addition, we also discuss work that has used noise exposure or pharmacological mimic of noise-induced excitotoxicity in zebrafish to define cellular mechanisms underlying noise-induced hair-cell damage and synapse loss. Lastly, we highlight how future studies in zebrafish could enhance our understanding of the pathological processes underlying synapse loss in both genetic and acquired auditory synaptopathy. This knowledge is critical in order to develop therapies that protect or repair auditory synaptic contacts.
Collapse
Affiliation(s)
- Katie S. Kindt
- Section on Sensory Cell Development and Function, NIDCD/National Institutes of Health, Bethesda, MD, United States
| | - Lavinia Sheets
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
41
|
Larval Zebrafish Lateral Line as a Model for Acoustic Trauma. eNeuro 2018; 5:eN-NWR-0206-18. [PMID: 30225343 PMCID: PMC6140105 DOI: 10.1523/eneuro.0206-18.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 07/25/2018] [Accepted: 08/08/2018] [Indexed: 12/14/2022] Open
Abstract
Excessive noise exposure damages sensory hair cells, leading to permanent hearing loss. Zebrafish are a highly tractable model that have advanced our understanding of drug-induced hair cell death, yet no comparable model exists for noise exposure research. We demonstrate the utility of zebrafish as model to increase understanding of hair cell damage from acoustic trauma and develop protective therapies. We created an acoustic trauma system using underwater cavitation to stimulate lateral line hair cells. We found that acoustic stimulation resulted in exposure time- and intensity-dependent lateral line and saccular hair cell damage that is maximal at 48–72 h post-trauma. The number of TUNEL+ lateral line hair cells increased 72 h post-exposure, whereas no increase was observed in TUNEL+ supporting cells, demonstrating that acoustic stimulation causes hair cell-specific damage. Lateral line hair cells damaged by acoustic stimulation regenerate within 3 d, consistent with prior regeneration studies utilizing ototoxic drugs. Acoustic stimulation-induced hair cell damage is attenuated by pharmacological inhibition of protein synthesis or caspase activation, suggesting a requirement for translation and activation of apoptotic signaling cascades. Surviving hair cells exposed to acoustic stimulation showed signs of synaptopathy, consistent with mammalian studies. Finally, we demonstrate the feasibility of this platform to identify compounds that prevent acoustic trauma by screening a small redox library for protective compounds. Our data suggest that acoustic stimulation results in lateral line hair cell damage consistent with acoustic trauma research in mammals, providing a highly tractable model for high-throughput genetic and drug discovery studies.
Collapse
|
42
|
Zhang L, Engler S, Koepcke L, Steenken F, Köppl C. Concurrent gradients of ribbon volume and AMPA-receptor patch volume in cochlear afferent synapses on gerbil inner hair cells. Hear Res 2018; 364:81-89. [DOI: 10.1016/j.heares.2018.03.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 02/26/2018] [Accepted: 03/28/2018] [Indexed: 11/16/2022]
|
43
|
Ji YR, Warrier S, Jiang T, Wu DK, Kindt KS. Directional selectivity of afferent neurons in zebrafish neuromasts is regulated by Emx2 in presynaptic hair cells. eLife 2018; 7:35796. [PMID: 29671737 PMCID: PMC5935481 DOI: 10.7554/elife.35796] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 04/18/2018] [Indexed: 11/23/2022] Open
Abstract
The orientation of hair bundles on top of sensory hair cells (HCs) in neuromasts of the lateral line system allows fish to detect direction of water flow. Each neuromast shows hair bundles arranged in two opposing directions and each afferent neuron innervates only HCs of the same orientation. Previously, we showed that this opposition is established by expression of Emx2 in half of the HCs, where it mediates hair bundle reversal (Jiang et al., 2017). Here, we show that Emx2 also regulates neuronal selection: afferent neurons innervate either Emx2-positive or negative HCs. In emx2 knockout and gain-of-function neuromasts, all HCs are unidirectional and the innervation patterns and physiological responses of the afferent neurons are dependent on the presence or absence of Emx2. Our results indicate that Emx2 mediates the directional selectivity of neuromasts by two distinct processes: regulating hair bundle orientation in HCs and selecting afferent neuronal targets.
Collapse
Affiliation(s)
- Young Rae Ji
- Section on Sensory Cell Regeneration and Development, Laboratory of Molecular Biology, Bethesda, United States
| | - Sunita Warrier
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, United States
| | - Tao Jiang
- Section on Sensory Cell Regeneration and Development, Laboratory of Molecular Biology, Bethesda, United States
| | - Doris K Wu
- Section on Sensory Cell Regeneration and Development, Laboratory of Molecular Biology, Bethesda, United States
| | - Katie S Kindt
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, United States
| |
Collapse
|
44
|
Zhang Q, Li S, Wong HTC, He XJ, Beirl A, Petralia RS, Wang YX, Kindt KS. Synaptically silent sensory hair cells in zebrafish are recruited after damage. Nat Commun 2018; 9:1388. [PMID: 29643351 PMCID: PMC5895622 DOI: 10.1038/s41467-018-03806-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 03/09/2018] [Indexed: 01/31/2023] Open
Abstract
Analysis of mechanotransduction among ensembles of sensory hair cells in vivo is challenging in many species. To overcome this challenge, we used optical indicators to investigate mechanotransduction among collections of hair cells in intact zebrafish. Our imaging reveals a previously undiscovered disconnect between hair-cell mechanosensation and synaptic transmission. We show that saturating mechanical stimuli able to open mechanically gated channels are unexpectedly insufficient to evoke vesicle fusion in the majority of hair cells. Although synaptically silent, latent hair cells can be rapidly recruited after damage, demonstrating that they are synaptically competent. Therefore synaptically silent hair cells may be an important reserve that acts to maintain sensory function. Our results demonstrate a previously unidentified level of complexity in sculpting sensory transmission from the periphery.
Collapse
Affiliation(s)
- Qiuxiang Zhang
- Section on Sensory Cell Development and Function, NIDCD/National Institutes of Health, Bethesda, MD, 20892, USA
| | - Suna Li
- Section on Sensory Cell Development and Function, NIDCD/National Institutes of Health, Bethesda, MD, 20892, USA
| | - Hiu-Tung C Wong
- Section on Sensory Cell Development and Function, NIDCD/National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xinyi J He
- Section on Sensory Cell Development and Function, NIDCD/National Institutes of Health, Bethesda, MD, 20892, USA
| | - Alisha Beirl
- Section on Sensory Cell Development and Function, NIDCD/National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ronald S Petralia
- Advanced Imaging Core, NIDCD/National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ya-Xian Wang
- Advanced Imaging Core, NIDCD/National Institutes of Health, Bethesda, MD, 20892, USA
| | - Katie S Kindt
- Section on Sensory Cell Development and Function, NIDCD/National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
45
|
Chen Z, Chou SW, McDermott BM. Ribeye protein is intrinsically dynamic but is stabilized in the context of the ribbon synapse. J Physiol 2018; 596:409-421. [PMID: 29086422 DOI: 10.1113/jp271215] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 10/26/2017] [Indexed: 01/16/2023] Open
Abstract
KEY POINTS The synaptic ribbon is an organelle that coordinates rapid and sustained vesicle release to enable hearing and balance. Ribeye a and b proteins are major constituents of the synaptic ribbon in hair cells. In this study, we use optically clear transgenic zebrafish to examine the potential dynamics of ribeye proteins in vivo. We demonstrate that ribeye proteins are inherently dynamic but are stabilized at the ribbons of hair cells in the ear and the lateral line system. ABSTRACT Ribeye protein is a major constituent of the synaptic ribbon, an organelle that coordinates rapid and sustained vesicle release to enable hearing and balance. The ribbon is considered to be a stable structure. However, under certain physiological conditions such as acoustic overexposure that results in temporary noise-induced hearing loss or perturbations of ion channels, ribbons may change shape or vanish altogether, suggesting greater plasticity than previously appreciated. The dynamic properties of ribeye proteins are unknown. Here we use transgenesis and imaging to explore the behaviours of ribeye proteins within the ribbon and also their intrinsic properties outside the context of the ribbon synapse in a control cell type, the skin cell. By fluorescence recovery after photobleaching (FRAP) on transgenic zebrafish larvae, we test whether ribeye proteins are dynamic in vivo in real time. In the skin, a cell type devoid of synaptic contacts, Ribeye a-mCherry exchanges with ribbon-like structures on a time scale of minutes (t1/2 = 3.2 min). In contrast, Ribeye a of the ear and lateral line and Ribeye b of the lateral line each exchange at ribbons of hair cells an order of magnitude slower (t1/2 of 125.6 min, 107.0 min and 95.3 min, respectively) than Ribeye a of the skin. These basal exchange rates suggest that long-term ribbon presence may require ribeye renewal. Our studies demonstrate that ribeye proteins are inherently dynamic but are stabilized at the ribbons of sensory cells in vivo.
Collapse
Affiliation(s)
- Zongwei Chen
- Department of Otolaryngology-Head and Neck Surgery, Case Western Reserve University, Cleveland, OH, 44106, USA.,Department of Biology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Shih-Wei Chou
- Department of Otolaryngology-Head and Neck Surgery, Case Western Reserve University, Cleveland, OH, 44106, USA.,Department of Biology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Brian M McDermott
- Department of Otolaryngology-Head and Neck Surgery, Case Western Reserve University, Cleveland, OH, 44106, USA.,Department of Biology, Case Western Reserve University, Cleveland, OH, 44106, USA.,Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA.,Department of Neurosciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| |
Collapse
|
46
|
Jean P, Lopez de la Morena D, Michanski S, Jaime Tobón LM, Chakrabarti R, Picher MM, Neef J, Jung S, Gültas M, Maxeiner S, Neef A, Wichmann C, Strenzke N, Grabner C, Moser T. The synaptic ribbon is critical for sound encoding at high rates and with temporal precision. eLife 2018; 7:29275. [PMID: 29328020 PMCID: PMC5794258 DOI: 10.7554/elife.29275] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/19/2017] [Indexed: 11/30/2022] Open
Abstract
We studied the role of the synaptic ribbon for sound encoding at the synapses between inner hair cells (IHCs) and spiral ganglion neurons (SGNs) in mice lacking RIBEYE (RBEKO/KO). Electron and immunofluorescence microscopy revealed a lack of synaptic ribbons and an assembly of several small active zones (AZs) at each synaptic contact. Spontaneous and sound-evoked firing rates of SGNs and their compound action potential were reduced, indicating impaired transmission at ribbonless IHC-SGN synapses. The temporal precision of sound encoding was impaired and the recovery of SGN-firing from adaptation indicated slowed synaptic vesicle (SV) replenishment. Activation of Ca2+-channels was shifted to more depolarized potentials and exocytosis was reduced for weak depolarizations. Presynaptic Ca2+-signals showed a broader spread, compatible with the altered Ca2+-channel clustering observed by super-resolution immunofluorescence microscopy. We postulate that RIBEYE disruption is partially compensated by multi-AZ organization. The remaining synaptic deficit indicates ribbon function in SV-replenishment and Ca2+-channel regulation. Our sense of hearing relies on our ears quickly and tirelessly processing information in a precise manner. Sounds cause vibrations in a part of the inner ear called the cochlea. Inside the cochlea, the vibrations move hair-like structures on sensory cells that translate these movements into electrical signals. These hair cells are connected to specialized nerve cells that relay the signals to the brain, which then interprets them as sounds. Hair cells communicate with the specialized nerve cells via connections known as chemical synapses. This means that the electrical signals in the hair cell activate channel proteins that allow calcium ions to flow in. This in turn triggers membrane-bound packages called vesicles inside the hair cell to fuse with its surface membrane and release their contents to the outside. The contents, namely chemicals called neurotransmitters, then travels across the space between the cells, relaying the signal to the nerve cell. The junctions between the hair cells and the nerve cells are more specifically known as ribbon synapses. This is because they have a ribbon-like structure that appears to tether a halo of vesicles close to the active zone where neurotransmitters are released. However, the exact role of this synaptic ribbon has remained mysterious despite decades of study. The ribbon is mainly composed of a protein called Ribeye, and now Jean, Lopez de la Morena, Michanski, Jaime Tobón et al. show that mutant mice that lack this protein do not have any ribbons at their “ribbon synapses”. Hair cells without synaptic ribbons are less able to timely and reliably send signals to the nerve cells, most likely because they cannot replenish the vesicles at the synapse quickly enough. Further analysis showed that the synaptic ribbon also helps to regulate the calcium channels at the synapse, which is important for linking the electrical signals in the hair cell to the release of the neurotransmitters. Jean et al. also saw that hair cells without ribbons reorganize their synapses to form multiple active zones that could transfer neurotransmitter to the nerve cells. This could partially compensate for the loss of the ribbons, meaning the impact of their loss may have been underestimated. Future studies could explore this by eliminating the Ribeye protein only after the ribbon synapses are fully formed. These findings may help scientists to better understand deafness and other hearing disorders in humans. They will also be of interest to neuroscientists who research synapses, hearing and other sensory processes.
Collapse
Affiliation(s)
- Philippe Jean
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Collaborative Research Center, University of Göttingen, Göttingen, Germany.,Göttingen Graduate School for Neurosciences and Molecular Biosciences, University of Göttingen, Göttingen, Germany.,InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
| | - David Lopez de la Morena
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Göttingen Graduate School for Neurosciences and Molecular Biosciences, University of Göttingen, Göttingen, Germany.,InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
| | - Susann Michanski
- Collaborative Research Center, University of Göttingen, Göttingen, Germany.,InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany.,Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany.,Institute for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Lina María Jaime Tobón
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Collaborative Research Center, University of Göttingen, Göttingen, Germany.,Göttingen Graduate School for Neurosciences and Molecular Biosciences, University of Göttingen, Göttingen, Germany.,InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany.,Synaptic Nanophysiology Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.,Auditory Neuroscience Group, Max Planck Institute for Experimental Medicine, Göttingen, Germany
| | - Rituparna Chakrabarti
- Collaborative Research Center, University of Göttingen, Göttingen, Germany.,Göttingen Graduate School for Neurosciences and Molecular Biosciences, University of Göttingen, Göttingen, Germany.,InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany.,Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany.,Institute for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Maria Magdalena Picher
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Collaborative Research Center, University of Göttingen, Göttingen, Germany.,InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
| | - Jakob Neef
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Collaborative Research Center, University of Göttingen, Göttingen, Germany.,InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany.,Synaptic Nanophysiology Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.,Auditory Neuroscience Group, Max Planck Institute for Experimental Medicine, Göttingen, Germany
| | - SangYong Jung
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany.,Neuro Modulation and Neuro Circuitry Group, Singapore Bioimaging Consortium (SBIC), Biomedical Sciences Institutes, Singapore, Singapore
| | - Mehmet Gültas
- Department of Breeding Informatics, Georg-August-University Göttingen, Göttingen, Germany
| | - Stephan Maxeiner
- Institute for Anatomy and Cell Biology, University of the Saarland, Homburg, Germany
| | - Andreas Neef
- Bernstein Group Biophysics of Neural Computation, Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
| | - Carolin Wichmann
- Collaborative Research Center, University of Göttingen, Göttingen, Germany.,InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany.,Molecular Architecture of Synapses Group, Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany.,Institute for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Nicola Strenzke
- Collaborative Research Center, University of Göttingen, Göttingen, Germany.,InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany.,Auditory Systems Physiology Group, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
| | - Chad Grabner
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany.,Synaptic Nanophysiology Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany.,Collaborative Research Center, University of Göttingen, Göttingen, Germany.,InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany.,Synaptic Nanophysiology Group, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany.,Auditory Neuroscience Group, Max Planck Institute for Experimental Medicine, Göttingen, Germany.,Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
47
|
Abstract
Our ears are remarkable sensory organs, providing the important senses of balance and hearing. The complex structure of the inner ear, or 'labyrinth', along with the assorted neuroepithelia, have evolved to detect head movements and sounds with impressive sensitivity. The rub is that the inner ear is highly vulnerable to genetic lesions and environmental insults. According to National Institute of Health estimates, hearing loss is one of the most commonly inherited or acquired sensorineural diseases. To understand the causes of deafness and balance disorders, it is imperative to understand the underlying biology of the inner ear, especially the inner workings of the sensory receptors. These receptors, which are termed hair cells, are particularly susceptible to genetic mutations - more than two dozen genes are associated with defects in this cell type in humans. Over the past decade, a substantial amount of progress has been made in working out the molecular basis of hair-cell function using vertebrate animal models. Given the transparency of the inner ear and the genetic tools that are available, zebrafish have become an increasingly popular animal model for the study of deafness and vestibular dysfunction. Mutagenesis screens for larval defects in hearing and balance have been fruitful in finding key components, many of which have been implicated in human deafness. This review will focus on the genes that are required for hair-cell function in zebrafish, with a particular emphasis on mechanotransduction. In addition, the generation of new tools available for the characterization of zebrafish hair-cell mutants will be discussed.
Collapse
Affiliation(s)
- Teresa Nicolson
- Oregon Hearing Research Center and the Vollum Institute, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, Tel: 503-494-3693,
| |
Collapse
|
48
|
In Vivo Ribbon Mobility and Turnover of Ribeye at Zebrafish Hair Cell Synapses. Sci Rep 2017; 7:7467. [PMID: 28785118 PMCID: PMC5547071 DOI: 10.1038/s41598-017-07940-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 07/03/2017] [Indexed: 11/08/2022] Open
Abstract
Ribbons are presynaptic structures that mediate synaptic vesicle release in some sensory cells of the auditory and visual systems. Although composed predominately of the protein Ribeye, very little is known about the structural dynamics of ribbons. Here we describe the in vivo mobility and turnover of Ribeye at hair cell ribbon synapses by monitoring fluorescence recovery after photobleaching (FRAP) in transgenic zebrafish with GFP-tagged Ribeye. We show that Ribeye can exchange between halves of a ribbon within ~1 minute in a manner that is consistent with a simple diffusion mechanism. In contrast, exchange of Ribeye between other ribbons via the cell's cytoplasm takes several hours.
Collapse
|
49
|
Daniele LL, Emran F, Lobo GP, Gaivin RJ, Perkins BD. Mutation of wrb, a Component of the Guided Entry of Tail-Anchored Protein Pathway, Disrupts Photoreceptor Synapse Structure and Function. Invest Ophthalmol Vis Sci 2017; 57:2942-54. [PMID: 27273592 PMCID: PMC4898200 DOI: 10.1167/iovs.15-18996] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Tail-anchored (TA) proteins contain a single hydrophobic domain at the C-terminus and are posttranslationally inserted into the ER membrane via the GET (guided entry of tail-anchored proteins) pathway. The role of the GET pathway in photoreceptors is unexplored. The goal of this study was to characterize the zebrafish pinball wizard mutant, which disrupts Wrb, a core component of the GET pathway. METHODS Electroretinography, optokinetic response measurements (OKR), immunohistochemistry, and electron microscopy analyses were employed to assess ribbon synapse function, protein expression, and ultrastructure in 5-day-old zebrafish larvae. Expression of wrb was investigated with real-time qRT-PCR and in situ hybridization. RESULTS Mutation of wrb abolished the OKR and greatly diminished the ERG b-wave, but not the a-wave. Ribeye and SV2 were partially mislocalized in both photoreceptors and hair cells of wrb mutants. Fewer contacts were seen between photoreceptors and bipolar cells in wrb-/- mutants. Expression of wrb was observed throughout the nervous system and Wrb localized to the ER and synaptic region of photoreceptors. Morpholino knockdown of the cytosolic ATPase trc40, which targets TA proteins to the ER, also diminished the OKR. Overexpression of wrb fully restored contrast sensitivity in mutants, while overexpression of mutant wrbR73A, which cannot bind Trc40, did not. CONCLUSIONS Proteins Wrb and Trc40 are required for synaptic transmission between photoreceptors and bipolar cells, indicating that TA protein insertion by the TRC pathway is a critical step in ribbon synapse assembly and function.
Collapse
Affiliation(s)
- Lauren L Daniele
- Department of Ophthalmic Research Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, United States
| | - Farida Emran
- Centre for Research in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Glenn P Lobo
- Department of Ophthalmic Research Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, United States
| | - Robert J Gaivin
- Department of Ophthalmic Research Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, United States
| | - Brian D Perkins
- Department of Ophthalmic Research Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, United States
| |
Collapse
|
50
|
Enlargement of Ribbons in Zebrafish Hair Cells Increases Calcium Currents But Disrupts Afferent Spontaneous Activity and Timing of Stimulus Onset. J Neurosci 2017; 37:6299-6313. [PMID: 28546313 PMCID: PMC5490065 DOI: 10.1523/jneurosci.2878-16.2017] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 04/26/2017] [Accepted: 04/27/2017] [Indexed: 12/26/2022] Open
Abstract
In sensory hair cells of auditory and vestibular organs, the ribbon synapse is required for the precise encoding of a wide range of complex stimuli. Hair cells have a unique presynaptic structure, the synaptic ribbon, which organizes both synaptic vesicles and calcium channels at the active zone. Previous work has shown that hair-cell ribbon size is correlated with differences in postsynaptic activity. However, additional variability in postsynapse size presents a challenge to determining the specific role of ribbon size in sensory encoding. To selectively assess the impact of ribbon size on synapse function, we examined hair cells in transgenic zebrafish that have enlarged ribbons, without postsynaptic alterations. Morphologically, we found that enlarged ribbons had more associated vesicles and reduced presynaptic calcium-channel clustering. Functionally, hair cells with enlarged ribbons had larger global and ribbon-localized calcium currents. Afferent neuron recordings revealed that hair cells with enlarged ribbons resulted in reduced spontaneous spike rates. Additionally, despite larger presynaptic calcium signals, we observed fewer evoked spikes with longer latencies from stimulus onset. Together, our work indicates that hair-cell ribbon size influences the spontaneous spiking and the precise encoding of stimulus onset in afferent neurons. SIGNIFICANCE STATEMENT Numerous studies support that hair-cell ribbon size corresponds with functional sensitivity differences in afferent neurons and, in the case of inner hair cells of the cochlea, vulnerability to damage from noise trauma. Yet it is unclear whether ribbon size directly influences sensory encoding. Our study reveals that ribbon enlargement results in increased ribbon-localized calcium signals, yet reduces afferent spontaneous activity and disrupts the timing of stimulus onset, a distinct aspect of auditory and vestibular encoding. These observations suggest that varying ribbon size alone can influence sensory encoding, and give further insight into how hair cells transduce signals that cover a wide dynamic range of stimuli.
Collapse
|