1
|
Vecchi JT, Rhomberg M, Allan Guymon C, Hansen MR. Inositol trisphosphate and ryanodine receptor signaling distinctly regulate neurite pathfinding in response to engineered micropatterned surfaces. PLoS One 2024; 19:e0308389. [PMID: 39236043 PMCID: PMC11376539 DOI: 10.1371/journal.pone.0308389] [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: 03/25/2024] [Accepted: 06/26/2024] [Indexed: 09/07/2024] Open
Abstract
Micro and nanoscale patterning of surface features and biochemical cues have emerged as tools to precisely direct neurite growth into close proximity with next generation neural prosthesis electrodes. Biophysical cues can exert greater influence on neurite pathfinding compared to the more well studied biochemical cues; yet the signaling events underlying the ability of growth cones to respond to these microfeatures remain obscure. Intracellular Ca2+ signaling plays a critical role in how a growth cone senses and grows in response to various cues (biophysical features, repulsive peptides, chemo-attractive gradients). Here, we investigate the role of inositol triphosphate (IP3) and ryanodine-sensitive receptor (RyR) signaling as sensory neurons (spiral ganglion neurons, SGNs, and dorsal root ganglion neurons, DRGNs) pathfind in response to micropatterned substrates of varied geometries. We find that IP3 and RyR signaling act in the growth cone as they navigate biophysical cues and enable proper guidance to biophysical, chemo-permissive, and chemo-repulsive micropatterns. In response to complex micropatterned geometries, RyR signaling appears to halt growth in response to both topographical features and chemo-repulsive cues. IP3 signaling appears to play a more complex role, as growth cones appear to sense the microfeatures in the presence of xestospongin C but are unable to coordinate turning in response to them. Overall, key Ca2+ signaling elements, IP3 and RyR, are found to be essential for SGNs to pathfind in response to engineered biophysical and biochemical cues. These findings inform efforts to precisely guide neurite regeneration for improved neural prosthesis function, including cochlear implants.
Collapse
Affiliation(s)
- Joseph T Vecchi
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, Iowa City, IA, United States of America
- Department of Otolaryngology Head-Neck Surgery, Carver College of Medicine, Iowa City, IA, United States of America
| | - Madeline Rhomberg
- Department of Otolaryngology Head-Neck Surgery, Carver College of Medicine, Iowa City, IA, United States of America
| | - C Allan Guymon
- Department of Chemical Engineering, University of Iowa, Iowa City, IA, United States of America
| | - Marlan R Hansen
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, Iowa City, IA, United States of America
- Department of Otolaryngology Head-Neck Surgery, Carver College of Medicine, Iowa City, IA, United States of America
| |
Collapse
|
2
|
Kim J, Martinez E, Qiu J, Zhouli Ni J, Kwan KY. Chromatin remodeling protein CHD4 regulates axon guidance of spiral ganglion neurons in developing cochlea. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578202. [PMID: 38352369 PMCID: PMC10862897 DOI: 10.1101/2024.01.31.578202] [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: 02/22/2024]
Abstract
The chromodomain helicase binding protein 4 (CHD4) is an ATP-dependent chromatin remodeler. De-novo pathogenic variants of CHD4 cause Sifrim-Hitz-Weiss syndrome (SIHIWES). Patients with SIHIWES show delayed development, intellectual disability, facial dysmorphism, and hearing loss. Many cochlear cell types, including spiral ganglion neurons (SGNs), express CHD4. SGNs are the primary afferent neurons that convey sound information from the cochlea, but the function of CHD4 in SGNs is unknown. We employed the Neurog1(Ngn1) CreERT2 Chd4 conditional knockout animals to delete Chd4 in SGNs. SGNs are classified as type I and type II neurons. SGNs lacking CHD4 showed abnormal fasciculation of type I neurons along with improper pathfinding of type II fibers. CHD4 binding to chromatin from immortalized multipotent otic progenitor-derived neurons was used to identify candidate target genes in SGNs. Gene ontology analysis of CHD4 target genes revealed cellular processes involved in axon guidance, axonal fasciculation, and ephrin receptor signaling pathway. We validated increased Epha4 transcripts in SGNs from Chd4 conditional knockout cochleae. The results suggest that CHD4 attenuates the transcription of axon guidance genes to form the stereotypic pattern of SGN peripheral projections. The results implicate epigenetic changes in circuit wiring by modulating axon guidance molecule expression and provide insights into neurodevelopmental diseases.
Collapse
Affiliation(s)
- Jihyun Kim
- Keck Center for Collaborative Neuroscience and Stem Cell Research Center, Rutgers University, Piscataway, NJ 08854, USA
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Edward Martinez
- Keck Center for Collaborative Neuroscience and Stem Cell Research Center, Rutgers University, Piscataway, NJ 08854, USA
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Jingyun Qiu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Julie Zhouli Ni
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA
| | - Kelvin Y. Kwan
- Keck Center for Collaborative Neuroscience and Stem Cell Research Center, Rutgers University, Piscataway, NJ 08854, USA
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| |
Collapse
|
3
|
Tomar M, Beros J, Meloni B, Rodger J. Interactions between Guidance Cues and Neuronal Activity: Therapeutic Insights from Mouse Models. Int J Mol Sci 2023; 24:ijms24086966. [PMID: 37108129 PMCID: PMC10138948 DOI: 10.3390/ijms24086966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023] Open
Abstract
Topographic mapping of neural circuits is fundamental in shaping the structural and functional organization of brain regions. This developmentally important process is crucial not only for the representation of different sensory inputs but also for their integration. Disruption of topographic organization has been associated with several neurodevelopmental disorders. The aim of this review is to highlight the mechanisms involved in creating and refining such well-defined maps in the brain with a focus on the Eph and ephrin families of axon guidance cues. We first describe the transgenic models where ephrin-A expression has been manipulated to understand the role of these guidance cues in defining topography in various sensory systems. We further describe the behavioral consequences of lacking ephrin-A guidance cues in these animal models. These studies have given us unexpected insight into how neuronal activity is equally important in refining neural circuits in different brain regions. We conclude the review by discussing studies that have used treatments such as repetitive transcranial magnetic stimulation (rTMS) to manipulate activity in the brain to compensate for the lack of guidance cues in ephrin-knockout animal models. We describe how rTMS could have therapeutic relevance in neurodevelopmental disorders with disrupted brain organization.
Collapse
Affiliation(s)
- Maitri Tomar
- School of Biological Sciences, The University of Western Australia, Crawley, WA 6009, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Jamie Beros
- School of Biological Sciences, The University of Western Australia, Crawley, WA 6009, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Bruno Meloni
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
- Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Crawley, WA 6009, Australia
- Department of Neurosurgery, Sir Charles Gairdner Hospital, QEII Medical Centre, Nedlands, WA 6009, Australia
| | - Jennifer Rodger
- School of Biological Sciences, The University of Western Australia, Crawley, WA 6009, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| |
Collapse
|
4
|
Cantu-Guerra HL, Papazian MR, Gorsky AL, Alekos NS, Caccavano A, Karagulyan N, Neef J, Vicini S, Moser T, Coate TM. Cochlear hair cell innervation is dependent on a modulatory function of Semaphorin-3A. Dev Dyn 2023; 252:124-144. [PMID: 36284453 PMCID: PMC9812910 DOI: 10.1002/dvdy.548] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/20/2022] [Accepted: 10/06/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Proper connectivity between type I spiral ganglion neurons (SGNs) and inner hair cells (IHCs) in the cochlea is necessary for conveying sound information to the brain in mammals. Previous studies have shown that type I SGNs are heterogeneous in form, function and synaptic location on IHCs, but factors controlling their patterns of connectivity are not well understood. RESULTS During development, cochlear supporting cells and SGNs express Semaphorin-3A (SEMA3A), a known axon guidance factor. Mice homozygous for a point mutation that attenuates normal SEMA3A repulsive activity (Sema3aK108N ) show cochleae with grossly normal patterns of IHC innervation. However, genetic sparse labeling and three-dimensional reconstructions of individual SGNs show that cochleae from Sema3aK108N mice lacked the normal synaptic distribution of type I SGNs. Additionally, Sema3aK108N cochleae show a disrupted distribution of GLUA2 postsynaptic patches around the IHCs. The addition of SEMA3A-Fc to postnatal cochleae led to increases in SGN branching, similar to the effects of inhibiting glutamate receptors. Ca2+ imaging studies show that SEMA3A-Fc decreases SGN activity. CONCLUSIONS Contrary to the canonical view of SEMA3A as a guidance ligand, our results suggest SEMA3A may regulate SGN excitability in the cochlea, which may influence the morphology and synaptic arrangement of type I SGNs.
Collapse
Affiliation(s)
- Homero L. Cantu-Guerra
- Department of Biology, Georgetown University, Washington,
District of Columbia, USA
- Interdisciplinary Program in Neuroscience, Georgetown
University, Washington, District of Columbia, USA
| | - Michael R. Papazian
- Department of Biology, Georgetown University, Washington,
District of Columbia, USA
| | - Anna L. Gorsky
- Department of Biology, Georgetown University, Washington,
District of Columbia, USA
| | - Nathalie S. Alekos
- Department of Biology, Georgetown University, Washington,
District of Columbia, USA
| | - Adam Caccavano
- Interdisciplinary Program in Neuroscience, Georgetown
University, Washington, District of Columbia, USA
- Department of Pharmacology, Georgetown University School of
Medicine, Washington, District of Columbia, USA
| | - Nare Karagulyan
- Institute for Auditory Neuroscience and InnerEarLab,
University Medical Center, and Auditory Neuroscience & Synaptic Nanophysiology
Group, Max Planck Institute for Multidisciplinary Sciences, and Cluster of
Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of
Excitable Cells” (MBExC), Göttingen, Germany
| | - Jakob Neef
- Institute for Auditory Neuroscience and InnerEarLab,
University Medical Center, and Auditory Neuroscience & Synaptic Nanophysiology
Group, Max Planck Institute for Multidisciplinary Sciences, and Cluster of
Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of
Excitable Cells” (MBExC), Göttingen, Germany
| | - Stefano Vicini
- Interdisciplinary Program in Neuroscience, Georgetown
University, Washington, District of Columbia, USA
- Department of Pharmacology, Georgetown University School of
Medicine, Washington, District of Columbia, USA
| | - Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLab,
University Medical Center, and Auditory Neuroscience & Synaptic Nanophysiology
Group, Max Planck Institute for Multidisciplinary Sciences, and Cluster of
Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of
Excitable Cells” (MBExC), Göttingen, Germany
| | - Thomas M. Coate
- Department of Biology, Georgetown University, Washington,
District of Columbia, USA
- Interdisciplinary Program in Neuroscience, Georgetown
University, Washington, District of Columbia, USA
| |
Collapse
|
5
|
Generation of innervated cochlear organoid recapitulates early development of auditory unit. Stem Cell Reports 2022; 18:319-336. [PMID: 36584686 PMCID: PMC9860115 DOI: 10.1016/j.stemcr.2022.11.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/30/2022] Open
Abstract
Functional cochlear hair cells (HCs) innervated by spiral ganglion neurons (SGNs) are essential for hearing, whereas robust models that recapitulate the peripheral auditory circuity are still lacking. Here, we developed cochlear organoids with functional peripheral auditory circuity in a staging three-dimensional (3D) co-culture system by initially reprogramming cochlear progenitor cells (CPCs) with increased proliferative potency that could be long-term expanded, then stepwise inducing the differentiation of cochlear HCs, as well as the outgrowth of neurites from SGNs. The function of HCs and synapses within organoids was confirmed by a series of morphological and electrophysiological evaluations. Single-cell mRNA sequencing revealed the differentiation trajectories of CPCs toward the major cochlear cell types and the dynamic gene expression during organoid HC development, which resembled the pattern of native HCs. We established the cochlear organoids with functional synapses for the first time, which provides a platform for deciphering the mechanisms of sensorineural hearing loss.
Collapse
|
6
|
Defourny J. Considering gene therapy to protect from X-linked deafness DFNX2 and associated neurodevelopmental disorders. IBRAIN 2022; 8:431-441. [PMID: 37786584 PMCID: PMC10529175 DOI: 10.1002/ibra.12068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 08/10/2022] [Accepted: 09/13/2022] [Indexed: 10/04/2023]
Abstract
Mutations and deletions in the gene or upstream of the gene encoding the POU3F4 transcription factor cause X-linked progressive deafness DFNX2 and additional neurodevelopmental disorders in humans. Hearing loss can be purely sensorineural or mixed, that is, with both conductive and sensorineural components. Affected males show anatomical abnormalities of the inner ear, which are jointly defined as incomplete partition type III. Current approaches to improve hearing and speech skills of DFNX2 patients do not seem to be fully effective. Owing to inner ear malformations, cochlear implantation is surgically difficult and may predispose towards severe complications. Even in cases where implantation is safely performed, hearing and speech outcomes remain highly variable among patients. Mouse models for DFNX2 deafness revealed that sensorineural loss could arise from a dysfunction of spiral ligament fibrocytes in the lateral wall of the cochlea, which leads to reduced endocochlear potential. Highly positive endocochlear potential is critical for sensory hair cell mechanotransduction and hearing. In this context, here, we propose to develop a therapeutic approach in male Pou3f4 -/y mice based on an adeno-associated viral (AAV) vector-mediated gene transfer in cochlear spiral ligament fibrocytes. Among a broad range of AAV vectors, AAV7 was found to show a strong tropism for the spiral ligament. Thus, we suggest that an AAV7-mediated delivery of Pou3f4 complementary DNA in the spiral ligament of Pou3f4 -/y mice could represent an attractive strategy to prevent fibrocyte degeneration and to restore normal cochlear functions and properties, including a positive endocochlear potential, before hearing loss progresses to profound deafness.
Collapse
Affiliation(s)
- Jean Defourny
- GIGA‐Neurosciences, Unit of Cell and Tissue BiologyUniversity of Liège, C.H.U. B36LiègeBelgium
| |
Collapse
|
7
|
Filova I, Pysanenko K, Tavakoli M, Vochyanova S, Dvorakova M, Bohuslavova R, Smolik O, Fabriciova V, Hrabalova P, Benesova S, Valihrach L, Cerny J, Yamoah EN, Syka J, Fritzsch B, Pavlinkova G. ISL1 is necessary for auditory neuron development and contributes toward tonotopic organization. Proc Natl Acad Sci U S A 2022; 119:e2207433119. [PMID: 36074819 PMCID: PMC9478650 DOI: 10.1073/pnas.2207433119] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 08/04/2022] [Indexed: 11/18/2022] Open
Abstract
A cardinal feature of the auditory pathway is frequency selectivity, represented in a tonotopic map from the cochlea to the cortex. The molecular determinants of the auditory frequency map are unknown. Here, we discovered that the transcription factor ISL1 regulates the molecular and cellular features of auditory neurons, including the formation of the spiral ganglion and peripheral and central processes that shape the tonotopic representation of the auditory map. We selectively knocked out Isl1 in auditory neurons using Neurod1Cre strategies. In the absence of Isl1, spiral ganglion neurons migrate into the central cochlea and beyond, and the cochlear wiring is profoundly reduced and disrupted. The central axons of Isl1 mutants lose their topographic projections and segregation at the cochlear nucleus. Transcriptome analysis of spiral ganglion neurons shows that Isl1 regulates neurogenesis, axonogenesis, migration, neurotransmission-related machinery, and synaptic communication patterns. We show that peripheral disorganization in the cochlea affects the physiological properties of hearing in the midbrain and auditory behavior. Surprisingly, auditory processing features are preserved despite the significant hearing impairment, revealing central auditory pathway resilience and plasticity in Isl1 mutant mice. Mutant mice have a reduced acoustic startle reflex, altered prepulse inhibition, and characteristics of compensatory neural hyperactivity centrally. Our findings show that ISL1 is one of the obligatory factors required to sculpt auditory structural and functional tonotopic maps. Still, upon Isl1 deletion, the ensuing central plasticity of the auditory pathway does not suffice to overcome developmentally induced peripheral dysfunction of the cochlea.
Collapse
Affiliation(s)
- Iva Filova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Kateryna Pysanenko
- Department of Auditory Neuroscience, Institute of Experimental Medicine Czech Academy of Sciences, 14220 Prague, Czechia
| | - Mitra Tavakoli
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Simona Vochyanova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Martina Dvorakova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Romana Bohuslavova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Ondrej Smolik
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Valeria Fabriciova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Petra Hrabalova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Sarka Benesova
- Laboratory of Gene Expression, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Lukas Valihrach
- Laboratory of Gene Expression, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| | - Jiri Cerny
- Laboratory of Light Microscopy, Institute of Molecular Genetics Czech Academy of Sciences, 14220 Prague, Czechia
| | - Ebenezer N. Yamoah
- Department of Physiology, School of Medicine, University of Nevada, Reno, NV 89557
| | - Josef Syka
- Department of Auditory Neuroscience, Institute of Experimental Medicine Czech Academy of Sciences, 14220 Prague, Czechia
| | - Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa City, IA 52242-1324
- Department of Otolaryngology, University of Iowa, Iowa City, IA 52242-1324
| | - Gabriela Pavlinkova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology Czech Academy of Sciences, 25250 Vestec, Czechia
| |
Collapse
|
8
|
Petitpré C, Faure L, Uhl P, Fontanet P, Filova I, Pavlinkova G, Adameyko I, Hadjab S, Lallemend F. Single-cell RNA-sequencing analysis of the developing mouse inner ear identifies molecular logic of auditory neuron diversification. Nat Commun 2022; 13:3878. [PMID: 35790771 PMCID: PMC9256748 DOI: 10.1038/s41467-022-31580-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 06/21/2022] [Indexed: 11/08/2022] Open
Abstract
Different types of spiral ganglion neurons (SGNs) are essential for auditory perception by transmitting complex auditory information from hair cells (HCs) to the brain. Here, we use deep, single cell transcriptomics to study the molecular mechanisms that govern their identity and organization in mice. We identify a core set of temporally patterned genes and gene regulatory networks that may contribute to the diversification of SGNs through sequential binary decisions and demonstrate a role for NEUROD1 in driving specification of a Ic-SGN phenotype. We also find that each trajectory of the decision tree is defined by initial co-expression of alternative subtype molecular controls followed by gradual shifts toward cell fate resolution. Finally, analysis of both developing SGN and HC types reveals cell-cell signaling potentially playing a role in the differentiation of SGNs. Our results indicate that SGN identities are drafted prior to birth and reveal molecular principles that shape their differentiation and will facilitate studies of their development, physiology, and dysfunction.
Collapse
Affiliation(s)
- Charles Petitpré
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Louis Faure
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, 1090, Vienna, Austria
| | - Phoebe Uhl
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Paula Fontanet
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Iva Filova
- Institute of Biotechnology CAS, 25250, Vestec, Czech Republic
| | | | - Igor Adameyko
- Department of Neuroimmunology, Center for Brain Research, Medical University Vienna, 1090, Vienna, Austria
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Saida Hadjab
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Francois Lallemend
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
- Ming-Wai Lau Centre for Reparative Medicine, Stockholm Node, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
9
|
Bartolomé A, Suda N, Yu J, Zhu C, Son J, Ding H, Califano A, Accili D, Pajvani UB. Notch-mediated Ephrin signaling disrupts islet architecture and β cell function. JCI Insight 2022; 7:157694. [PMID: 35167496 PMCID: PMC8986078 DOI: 10.1172/jci.insight.157694] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/09/2022] [Indexed: 11/23/2022] Open
Abstract
Altered islet architecture is associated with β cell dysfunction and type 2 diabetes (T2D) progression, but molecular effectors of islet spatial organization remain mostly unknown. Although Notch signaling is known to regulate pancreatic development, we observed “reactivated” β cell Notch activity in obese mouse models. To test the repercussions and reversibility of Notch effects, we generated doxycycline-dependent, β cell–specific Notch gain-of-function mice. As predicted, we found that Notch activation in postnatal β cells impaired glucose-stimulated insulin secretion and glucose intolerance, but we observed a surprising remnant glucose intolerance after doxycycline withdrawal and cessation of Notch activity, associated with a marked disruption of normal islet architecture. Transcriptomic screening of Notch-active islets revealed increased Ephrin signaling. Commensurately, exposure to Ephrin ligands increased β cell repulsion and impaired murine and human pseudoislet formation. Consistent with our mouse data, Notch and Ephrin signaling were increased in metabolically inflexible β cells in patients with T2D. These studies suggest that β cell Notch/Ephrin signaling can permanently alter islet architecture during a morphogenetic window in early life.
Collapse
Affiliation(s)
- Alberto Bartolomé
- Departamento de Fisiopatología Endocrina y del Sistema Nervioso, IIBm Alberto Sols (CSIC/UAM), Madrid, Spain
| | - Nina Suda
- Department of Medicine, Columbia University, New York, United States of America
| | - Junjie Yu
- Department of Medicine, Columbia University, New York, United States of America
| | - Changyu Zhu
- Department of Medicine, Columbia University, New York, United States of America
| | - Jinsook Son
- Department of Medicine, Columbia University, New York, United States of America
| | - Hongxu Ding
- Systems Biology, Columbia University College of Physicians & Surgeons, New York, United States of America
| | - Andrea Califano
- Systems Biology, Columbia University College of Physicians & Surgeons, New York, United States of America
| | - Domenico Accili
- Department of Medicine, Columbia University, New York, United States of America
| | - Utpal B Pajvani
- Department of Medicine, Columbia University, New York, United States of America
| |
Collapse
|
10
|
Abstract
PURPOSE OF REVIEW We review recent progress in the characterization of spiral ganglion neurons (SGNs), the afferent neurons that transmit sound information from mechanosensory hair cells in the inner ear to the central nervous system. RECENT FINDINGS Single-cell ribonucleic acid sequencing studies of murine SGNs have demonstrated that SGNs consist of molecularly distinct subtypes. The molecularly defined SGN subtypes likely correspond to SGN subtypes previously identified on the basis of physiological properties, although this has not been experimentally demonstrated. Subtype maturation is completed postnatally in an activity-dependent manner and is impaired in several models of hearing loss. SUMMARY The recent molecular studies open new avenues to rigorously test whether SGN subtypes are important for the encoding of different sound features and if they show differential vulnerability to genetic factors and environmental insults. This could have important implications for the development of therapeutic strategies to treat hearing loss.
Collapse
Affiliation(s)
- Shuohao Sun
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | |
Collapse
|
11
|
Interaction of micropatterned topographical and biochemical cues to direct neurite growth from spiral ganglion neurons. Hear Res 2021; 409:108315. [PMID: 34343850 DOI: 10.1016/j.heares.2021.108315] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/07/2021] [Accepted: 07/12/2021] [Indexed: 01/01/2023]
Abstract
Functional outcomes with neural prosthetic devices, such as cochlear implants, are limited in part due to physical separation between the stimulating elements and the neurons they stimulate. One strategy to close this gap aims to precisely guide neurite regeneration to position the neurites in closer proximity to electrode arrays. Here, we explore the ability of micropatterned biochemical and topographic guidance cues, singly and in combination, to direct the growth of spiral ganglion neuron (SGN) neurites, the neurons targeted by cochlear implants. Photopolymerization of methacrylate monomers was used to form unidirectional topographical features of ridges and grooves in addition to multidirectional patterns with 90o angle turns. Microcontact printing was also used to create similar uni- and multi-directional patterns of peptides on polymer surfaces. Biochemical cues included peptides that facilitate (laminin, LN) or repel (EphA4-Fc) neurite growth. On flat surfaces, SGN neurites preferentially grew on LN-coated stripes and avoided EphA4-Fc-coated stripes. LN or EphA4-Fc was selectively adsorbed onto the ridges or grooves to test the neurite response to a combination of topographical and biochemical cues. Coating the ridges with EphA4-Fc and grooves with LN lead to enhanced SGN alignment to topographical patterns. Conversely, EphA4-Fc coating on the grooves or LN coating on the ridges tended to disrupt alignment to topographical patterns. SGN neurites respond to combinations of topographical and biochemical cues and surface patterning that leverages both cues enhance guided neurite growth.
Collapse
|
12
|
Koizumi Y, Mizutari K, Kawauchi S, Sato S, Shiotani A, Kakehata S. Y-27632, a ROCK inhibitor, improved laser-induced shock wave (LISW)-induced cochlear synaptopathy in mice. Mol Brain 2021; 14:105. [PMID: 34217338 PMCID: PMC8254252 DOI: 10.1186/s13041-021-00819-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/28/2021] [Indexed: 11/10/2022] Open
Abstract
Recently, a pathological condition called cochlear synaptopathy has been clarified, and as a disorder of the auditory nerve synapses that occurs prior to failure of hair cells, it has been recognized as a major cause of sensorineural hearing loss. However, cochlear synaptopathy is untreatable. Inhibition of rho-associated coiled-coil containing protein kinase (ROCK), a serine-threonine protein kinase, has been reported to have neuroprotective and regenerative effects on synaptic pathways in the nervous system, including those in the inner ear. We previously demonstrated the regenerative effect of the ROCK inhibitor, Y-27632, on an excitotoxic cochlear nerve damage model in vitro. In this study, we aimed to validate the effect of ROCK inhibition on mice with cochlear synaptopathy induced by laser-induced shock wave (LISW) in vivo. After the elevation of ROCK1/2 expression in the damaged cochlea was confirmed, we administered Y-27632 locally via the middle ear. The amplitude of wave I in the auditory brainstem response and the number of synapses in the Y-27632-treated cochlea increased significantly. These results clearly demonstrate that ROCK inhibition has a promising clinical application in the treatment of cochlear synaptopathy, which is the major pathology of sensorineural hearing loss.
Collapse
Affiliation(s)
- Yutaka Koizumi
- Department of Otolaryngology-Head and Neck Surgery, Yamagata University Faculty of Medicine, 2-2-2 Iida-Nishi, Yamagata, 990-9585, Japan
| | - Kunio Mizutari
- Department of Otolaryngology-Head and Neck Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan.
| | - Satoko Kawauchi
- Division of Bioinformation and Therapeutic Systems, National Defense Medical College Research Institute, Saitama, 359-8513, Japan
| | - Shunichi Sato
- Division of Bioinformation and Therapeutic Systems, National Defense Medical College Research Institute, Saitama, 359-8513, Japan
| | - Akihiro Shiotani
- Department of Otolaryngology-Head and Neck Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Seiji Kakehata
- Department of Otolaryngology-Head and Neck Surgery, Yamagata University Faculty of Medicine, 2-2-2 Iida-Nishi, Yamagata, 990-9585, Japan.
| |
Collapse
|
13
|
Elliott KL, Kersigo J, Lee JH, Jahan I, Pavlinkova G, Fritzsch B, Yamoah EN. Developmental Changes in Peripherin-eGFP Expression in Spiral Ganglion Neurons. Front Cell Neurosci 2021; 15:678113. [PMID: 34211371 PMCID: PMC8239239 DOI: 10.3389/fncel.2021.678113] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/26/2021] [Indexed: 11/13/2022] Open
Abstract
The two types of spiral ganglion neurons (SGNs), types I and II, innervate inner hair cells and outer hair cells, respectively, within the mammalian cochlea and send another process back to cochlear nuclei in the hindbrain. Studying these two neuronal types has been made easier with the identification of unique molecular markers. One of these markers, peripherin, was shown using antibodies to be present in all SGNs initially but becomes specific to type II SGNs during maturation. We used mice with fluorescently labeled peripherin (Prph-eGFP) to examine peripherin expression in SGNs during development and in aged mice. Using these mice, we confirm the initial expression of Prph-eGFP in both types I and II neurons and eventual restriction to only type II perikarya shortly after birth. However, while Prph-eGFP is uniquely expressed within type II cell bodies by P8, both types I and II peripheral and central processes continue to express Prph-eGFP for some time before becoming downregulated. Only at P30 was there selective type II Prph-eGFP expression in central but not peripheral processes. By 9 months, only the type II cell bodies and more distal central processes retain Prph-eGFP expression. Our results show that Prph-eGFP is a reliable marker for type II SGN cell bodies beyond P8; however, it is not generally a suitable marker for type II processes, except for central processes beyond P30. How the changes in Prph-eGFP expression relate to subsequent protein expression remains to be explored.
Collapse
Affiliation(s)
- Karen L Elliott
- Department of Biology, CLAS, The University of Iowa, Iowa City, IA, United States.,Department of Otolaryngology, CLAS, The University of Iowa, Iowa City, IA, United States
| | - Jennifer Kersigo
- Department of Biology, CLAS, The University of Iowa, Iowa City, IA, United States.,Department of Otolaryngology, CLAS, The University of Iowa, Iowa City, IA, United States
| | - Jeong Han Lee
- Department of Physiology, School of Medicine, University of Nevada, Reno, Reno, NV, United States
| | - Israt Jahan
- Department of Biology, CLAS, The University of Iowa, Iowa City, IA, United States.,Department of Otolaryngology, CLAS, The University of Iowa, Iowa City, IA, United States
| | | | - Bernd Fritzsch
- Department of Biology, CLAS, The University of Iowa, Iowa City, IA, United States.,Department of Otolaryngology, CLAS, The University of Iowa, Iowa City, IA, United States
| | - Ebenezer N Yamoah
- Department of Physiology, School of Medicine, University of Nevada, Reno, Reno, NV, United States
| |
Collapse
|
14
|
Defourny J, Audouard C, Davy A, Thiry M. Efnb2 haploinsufficiency induces early gap junction plaque disassembly and endocytosis in the cochlea. Brain Res Bull 2021; 174:153-160. [PMID: 34139316 DOI: 10.1016/j.brainresbull.2021.06.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 06/07/2021] [Accepted: 06/11/2021] [Indexed: 11/15/2022]
Abstract
Chromosome 13q deletions encompassing EFNB2, which encodes the transmembrane protein ephrin-B2, are likely to cause syndromic forms of sensorineural hearing loss of unclear origin. Thus, unravelling the pathogenic mechanisms could help to improve therapeutic strategies. In the cochlea, adjacent non-sensory epithelial cells are connected via gap junction channels, the activity of which is critical to maintain cochlear homeostasis. Here we show that ephrin-B2 promotes the assembly of connexin 30 (Cx30) gap junction plaques (GJPs) between adjacent non-sensory Deiters' cells. An in situ proximity ligation assay revealed that ephrin-B2 preferentially interacts with Cx30 in the periphery of the GJPs, i.e. where newly synthesized connexin hemichannels accrue to the GJP. Moreover, we observed that heterozygous mice encoding an Efnb2 null allele display excessive clathrin-mediated internalization of Cx30 GJPs in early postnatal stages. Finally, an in vitro organotypic assay revealed that ectopic activation of ephrin-B2 reverse signalling promotes the internalization of Cx30 GJPs. These data argue in favor of a cell-autonomous, Eph receptor-independent role of ephrin-B2 in the assembly of Cx30 GJPs. According to recent observations, early GJP degradation could certainly play a role in the pathogenic process leading to progressive sensorineural hearing loss due to Efnb2/EFNB2 haploinsufficiency.
Collapse
Affiliation(s)
- Jean Defourny
- GIGA-Neurosciences, Unit of Cell and Tissue Biology, University of Liège, CHU B36, 4000, Liège, Belgium.
| | - Christophe Audouard
- Center for Developmental Biology, Center for Integrative Biology, University of Toulouse, CNRS, UPS, 118 route de Narbonne, 31062, Toulouse, France
| | - Alice Davy
- Center for Developmental Biology, Center for Integrative Biology, University of Toulouse, CNRS, UPS, 118 route de Narbonne, 31062, Toulouse, France
| | - Marc Thiry
- GIGA-Neurosciences, Unit of Cell and Tissue Biology, University of Liège, CHU B36, 4000, Liège, Belgium
| |
Collapse
|
15
|
Sitko AA, Goodrich LV. Making sense of neural development by comparing wiring strategies for seeing and hearing. Science 2021; 371:eaaz6317. [PMID: 33414193 PMCID: PMC8034811 DOI: 10.1126/science.aaz6317] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The ability to perceive and interact with the world depends on a diverse array of neural circuits specialized for carrying out specific computations. Each circuit is assembled using a relatively limited number of molecules and common developmental steps, from cell fate specification to activity-dependent synaptic refinement. Given this shared toolkit, how do individual circuits acquire their characteristic properties? We explore this question by comparing development of the circuitry for seeing and hearing, highlighting a few examples where differences in each system's sensory demands necessitate different developmental strategies.
Collapse
Affiliation(s)
- A A Sitko
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - L V Goodrich
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Webber JL, Clancy JC, Zhou Y, Yraola N, Homma K, García-Añoveros J. Axodendritic versus axosomatic cochlear efferent termination is determined by afferent type in a hierarchical logic of circuit formation. SCIENCE ADVANCES 2021; 7:7/4/eabd8637. [PMID: 33523928 PMCID: PMC7817091 DOI: 10.1126/sciadv.abd8637] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 12/03/2020] [Indexed: 05/09/2023]
Abstract
Hearing involves a stereotyped neural network communicating cochlea and brain. How this sensorineural circuit assembles is largely unknown. The cochlea houses two types of mechanosensory hair cells differing in function (sound transmission versus amplification) and location (inner versus outer compartments). Inner (IHCs) and outer hair cells (OHCs) are each innervated by a distinct pair of afferent and efferent neurons: IHCs are contacted by type I afferents receiving axodendritic efferent contacts; OHCs are contacted by type II afferents and axosomatically terminating efferents. Using an Insm1 mouse mutant with IHCs in the position of OHCs, we discover a hierarchical sequence of instructions in which first IHCs attract, and OHCs repel, type I afferents; second, type II afferents innervate hair cells not contacted by type I afferents; and last, afferent fiber type determines if and how efferents innervate, whether axodendritically on the afferent, axosomatically on the hair cell, or not at all.
Collapse
Affiliation(s)
- Jemma L Webber
- Department of Anesthesiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - John C Clancy
- Department of Anesthesiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Yingjie Zhou
- Department of Anesthesiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Natalia Yraola
- Department of Anesthesiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Kazuaki Homma
- Department of Otolaryngology-Head and Neck Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- The Hugh Knowles Center for Clinical and Basic Science in Hearing and its Disorders, Northwestern University, Chicago, IL 60611, USA
| | - Jaime García-Añoveros
- Department of Anesthesiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
- The Hugh Knowles Center for Clinical and Basic Science in Hearing and its Disorders, Northwestern University, Chicago, IL 60611, USA
- Departments of Neurology and Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
17
|
Koizumi Y, Ito T, Mizutari K, Kakehata S. Regenerative Effect of a ROCK Inhibitor, Y-27632, on Excitotoxic Trauma in an Organotypic Culture of the Cochlea. Front Cell Neurosci 2020; 14:572434. [PMID: 33328888 PMCID: PMC7717995 DOI: 10.3389/fncel.2020.572434] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 10/26/2020] [Indexed: 11/28/2022] Open
Abstract
In the past, most inner ear diseases were thought to start with the impairment of the sensory epithelium of the cochlea before subsequently progressing to secondary neural degeneration. However, recent studies show that loss of primary synapses accompanied by excitotoxic degeneration of peripheral axons is likely to be the underlying pathology in sensorineural hearing loss. Rho-associated coiled-coil containing protein kinase (ROCK) inhibition has been reported to have neuroprotective and regenerative effects on synaptic pathways. Therefore, we analyzed the effect of ROCK inhibition using Y-27632 in a model of peripheral axonal damage in the spiral ganglion neurons created using the glutamate agonists, N-methyl-D-aspartate (NMDA) and kainic acid, to induce excitotoxic trauma in the explanted cochlea. The number of axons projecting to hair cells in the cochlea treated with Y-27632 was significantly greater than those in the cochlea treated only with NMDA + kainic acid. Furthermore, there was a significant increase in synapses between the spiral ganglion and the inner hair cells in the cochlea treated with Y-27632. The findings of this study suggest that ROCK inhibition could be a potential strategy for the regeneration of peripheral axons in the spiral ganglion and synapse formation in the inner hair cells of a cochlea that has sustained excitotoxic injury, which is one of the primary etiologies of inner ear disease.
Collapse
Affiliation(s)
- Yutaka Koizumi
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine, Yamagata University, Yamagata, Japan
| | - Tsukasa Ito
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine, Yamagata University, Yamagata, Japan
| | - Kunio Mizutari
- Department of Otolaryngology-Head and Neck Surgery, National Defense Medical College, Saitama, Japan
| | - Seiji Kakehata
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine, Yamagata University, Yamagata, Japan
| |
Collapse
|
18
|
Neuronal processes and glial precursors form a scaffold for wiring the developing mouse cochlea. Nat Commun 2020; 11:5866. [PMID: 33203842 PMCID: PMC7672226 DOI: 10.1038/s41467-020-19521-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 10/02/2020] [Indexed: 01/20/2023] Open
Abstract
In the developing nervous system, axons navigate through complex terrains that change depending on when and where outgrowth begins. For instance, in the developing cochlea, spiral ganglion neurons extend their peripheral processes through a growing and heterogeneous environment en route to their final targets, the hair cells. Although the basic principles of axon guidance are well established, it remains unclear how axons adjust strategies over time and space. Here, we show that neurons with different positions in the spiral ganglion employ different guidance mechanisms, with evidence for both glia-guided growth and fasciculation along a neuronal scaffold. Processes from neurons in the rear of the ganglion are more directed and grow faster than those from neurons at the border of the ganglion. Further, processes at the wavefront grow more efficiently when in contact with glial precursors growing ahead of them. These findings suggest a tiered mechanism for reliable axon guidance.
Collapse
|
19
|
Hosoya M, Fujioka M, Murayama AY, Okano H, Ogawa K. The common marmoset as suitable nonhuman alternative for the analysis of primate cochlear development. FEBS J 2020; 288:325-353. [PMID: 32323465 PMCID: PMC7818239 DOI: 10.1111/febs.15341] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/30/2020] [Accepted: 04/20/2020] [Indexed: 12/13/2022]
Abstract
Cochlear development is a complex process with precise spatiotemporal patterns. A detailed understanding of this process is important for studies of congenital hearing loss and regenerative medicine. However, much of our understanding of cochlear development is based on rodent models. Animal models that bridge the gap between humans and rodents are needed. In this study, we investigated the development of hearing organs in a small New World monkey species, the common marmoset (Callithrix jacchus). We describe the general stages of cochlear development in comparison with those of humans and mice. Moreover, we examined more than 25 proteins involved in cochlear development and found that expression patterns were generally conserved between rodents and primates. However, several proteins involved in supporting cell processes and neuronal development exhibited interspecific expression differences. Human fetal samples for studies of primate‐specific cochlear development are extremely rare, especially for late developmental stages. Our results support the use of the common marmoset as an effective alternative for analyses of primate cochlear development.
Collapse
Affiliation(s)
- Makoto Hosoya
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Masato Fujioka
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Ayako Y Murayama
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan.,Laboratory for Marmoset Neural Architecture, Center for Brain Science, RIKEN, Wako, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan.,Laboratory for Marmoset Neural Architecture, Center for Brain Science, RIKEN, Wako, Japan
| | - Kaoru Ogawa
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
20
|
Li M, Nishio SY, Naruse C, Riddell M, Sapski S, Katsuno T, Hikita T, Mizapourshafiyi F, Smith FM, Cooper LT, Lee MG, Asano M, Boettger T, Krueger M, Wietelmann A, Graumann J, Day BW, Boyd AW, Offermanns S, Kitajiri SI, Usami SI, Nakayama M. Digenic inheritance of mutations in EPHA2 and SLC26A4 in Pendred syndrome. Nat Commun 2020; 11:1343. [PMID: 32165640 PMCID: PMC7067772 DOI: 10.1038/s41467-020-15198-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 02/17/2020] [Indexed: 01/03/2023] Open
Abstract
Enlarged vestibular aqueduct (EVA) is one of the most commonly identified inner ear malformations in hearing loss patients including Pendred syndrome. While biallelic mutations of the SLC26A4 gene, encoding pendrin, causes non-syndromic hearing loss with EVA or Pendred syndrome, a considerable number of patients appear to carry mono-allelic mutation. This suggests faulty pendrin regulatory machinery results in hearing loss. Here we identify EPHA2 as another causative gene of Pendred syndrome with SLC26A4. EphA2 forms a protein complex with pendrin controlling pendrin localization, which is disrupted in some pathogenic forms of pendrin. Moreover, point mutations leading to amino acid substitution in the EPHA2 gene are identified from patients bearing mono-allelic mutation of SLC26A4. Ephrin-B2 binds to EphA2 triggering internalization with pendrin inducing EphA2 autophosphorylation weakly. The identified EphA2 mutants attenuate ephrin-B2- but not ephrin-A1-induced EphA2 internalization with pendrin. Our results uncover an unexpected role of the Eph/ephrin system in epithelial function. While biallelic mutations of the SLC26A4 gene cause non-syndromic hearing loss with enlarged vestibular aqueducts or Pendred syndrome, a considerable number of patients carry mono-allelic mutations. Here the authors identify EPHA2 as another causative gene of Pendred syndrome with SLC26A4.
Collapse
Affiliation(s)
- Mengnan Li
- Laboratory for Cell Polarity and Organogenesis, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, Philipps-Universität Marburg, Marburg, Germany
| | - Shin-Ya Nishio
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Chie Naruse
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Meghan Riddell
- Laboratory for Cell Polarity and Organogenesis, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Sabrina Sapski
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Tatsuya Katsuno
- Department of Otolaryngology - Head and Neck Surgery Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takao Hikita
- Laboratory for Cell Polarity and Organogenesis, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Fatemeh Mizapourshafiyi
- Laboratory for Cell Polarity and Organogenesis, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, Philipps-Universität Marburg, Marburg, Germany
| | - Fiona M Smith
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Leanne T Cooper
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Min Goo Lee
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea
| | - Masahide Asano
- Institute of Laboratory Animals, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Thomas Boettger
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Marcus Krueger
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Astrid Wietelmann
- MRI and µCT Service Group, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Johannes Graumann
- Scientific Service Group Biomolecular Mass Spectrometry Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site - Rhine-Main, Berlin, Germany
| | - Bryan W Day
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Andrew W Boyd
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Shin-Ichiro Kitajiri
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Shin-Ichi Usami
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Masanori Nakayama
- Laboratory for Cell Polarity and Organogenesis, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany. .,DFG Research Training Group, Membrane Plasticity in Tissue Development and Remodeling, GRK 2213, Philipps-Universität Marburg, Marburg, Germany. .,Kumamoto University International Research Center for Medical Scinece, Kumamoto, Japan.
| |
Collapse
|
21
|
Jung JS, Zhang KD, Wang Z, McMurray M, Tkaczuk A, Ogawa Y, Hertzano R, Coate TM. Semaphorin-5B Controls Spiral Ganglion Neuron Branch Refinement during Development. J Neurosci 2019; 39:6425-6438. [PMID: 31209173 PMCID: PMC6697390 DOI: 10.1523/jneurosci.0113-19.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 05/03/2019] [Accepted: 06/10/2019] [Indexed: 01/30/2023] Open
Abstract
During nervous system development, axons often undergo elaborate changes in branching patterns before circuits have achieved their mature patterns of innervation. In the auditory system, type I spiral ganglion neurons (SGNs) project their peripheral axons into the cochlear epithelium and then undergo a process of branch refinement before forming synapses with sensory hair cells. Here, we report that Semaphorin-5B (Sema5B) acts as an important mediator of this process. During cochlear development in mouse, immature hair cells express Sema5B, whereas the SGNs express both PlexinA1 and PlexinA3, which are known Sema5B receptors. In these studies, genetic sparse labeling and three-dimensional reconstruction techniques were leveraged to determine the morphologies of individual type I SGNs after manipulations of Sema5B signaling. Treating cultured mouse cochleae with Sema5B-Fc (to activate Plexin-As) led to type I SGNs with less numerous, but longer terminal branches. Conversely, cochleae from Sema5b knock-out mice showed type I SGNs with more numerous, but shorter terminal branches. In addition, conditional loss of Plxna1 in SGNs (using Bhlhb5Cre) led to increased type I SGN branching, suggesting that PlexinA1 normally responds to Sema5B in this process. In these studies, mice of either sex were used. The data presented here suggest that Sema5B-PlexinA1 signaling limits SGN terminal branch numbers without causing axonal repulsion, which is a role that distinguishes Sema5B from other Semaphorins in cochlear development.SIGNIFICANCE STATEMENT The sensorineural components of the cochlea include hair cells, which respond mechanically to sound waves, and afferent spiral ganglion neurons (SGNs), which respond to glutamate released by hair cells and transmit auditory information into the CNS. An important component of synapse formation in the cochlea is a process of SGN "debranching" whereby SGNs lose extraneous branches before developing unramified bouton endings that contact the hair cells. In this work, we have found that the transmembrane ligand Semaphorin-5B and its receptor PlexinA1 regulate the debranching process. The results in this report provide new knowledge regarding the molecular control of cochlear afferent innervation.
Collapse
Affiliation(s)
- Johnny S Jung
- Department of Biology, Georgetown University, Washington, DC 20007, and
| | - Kaidi D Zhang
- Department of Biology, Georgetown University, Washington, DC 20007, and
| | - Zhirong Wang
- Department of Biology, Georgetown University, Washington, DC 20007, and
| | - Mark McMurray
- Departments of Otorhinolaryngology Head and Neck Surgery
| | - Andrew Tkaczuk
- Departments of Otorhinolaryngology Head and Neck Surgery
| | - Yoko Ogawa
- Departments of Otorhinolaryngology Head and Neck Surgery
| | - Ronna Hertzano
- Departments of Otorhinolaryngology Head and Neck Surgery
- Anatomy and Neurobiology, and
- Institute for Genome Sciences, University of Maryland School of Medicine, University of Maryland, Baltimore, Maryland 21201
| | - Thomas M Coate
- Department of Biology, Georgetown University, Washington, DC 20007, and
| |
Collapse
|
22
|
Rué L, Oeckl P, Timmers M, Lenaerts A, van der Vos J, Smolders S, Poppe L, de Boer A, Van Den Bosch L, Van Damme P, Weishaupt JH, Ludolph AC, Otto M, Robberecht W, Lemmens R. Reduction of ephrin-A5 aggravates disease progression in amyotrophic lateral sclerosis. Acta Neuropathol Commun 2019; 7:114. [PMID: 31300041 PMCID: PMC6626434 DOI: 10.1186/s40478-019-0759-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 06/22/2019] [Indexed: 12/30/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease that affects motor neurons in the brainstem, spinal cord and motor cortex. ALS is characterized by genetic and clinical heterogeneity, suggesting the existence of genetic factors that modify the phenotypic expression of the disease. We previously identified the axonal guidance EphA4 receptor, member of the Eph-ephrin system, as an ALS disease-modifying factor. EphA4 genetic inhibition rescued the motor neuron phenotype in zebrafish and a rodent model of ALS. Preventing ligands from binding to the EphA4 receptor also successfully improved disease, suggesting a role for EphA4 ligands in ALS. One particular ligand, ephrin-A5, is upregulated in reactive astrocytes after acute neuronal injury and inhibits axonal regeneration. Moreover, it plays a role during development in the correct pathfinding of motor axons towards their target limb muscles. We hypothesized that a constitutive reduction of ephrin-A5 signalling would benefit disease progression in a rodent model for ALS. We discovered that in the spinal cord of control and symptomatic ALS mice ephrin-A5 was predominantly expressed in neurons. Surprisingly, reduction of ephrin-A5 levels in SOD1G93A mice accelerated disease progression and reduced survival without affecting disease onset, motor neuron numbers or innervated neuromuscular junctions in symptomatic mice. These findings suggest ephrin-A5 as a modifier of disease progression that might play a role in the later stages of the disease. Similarly, we identified a more aggressive disease progression in patients with lower ephrin-A5 protein levels in the cerebrospinal fluid without modifying disease onset. In summary, we identified reduced expression of ephrin-A5 to accelerate disease progression in a mouse model of ALS as well as in humans. Combined with our previous findings on the role of EphA4 in ALS our current data suggests different contribution for various members of the Eph-ephrin system in the pathophysiology of a motor neuron disease.
Collapse
|
23
|
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
|
24
|
Scott MK, Yue J, Biesemeier DJ, Lee JW, Fekete DM. Expression of class III Semaphorins and their receptors in the developing chicken (Gallus gallus) inner ear. J Comp Neurol 2019; 527:1196-1209. [PMID: 30520042 PMCID: PMC6401314 DOI: 10.1002/cne.24595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/15/2018] [Accepted: 11/23/2018] [Indexed: 12/22/2022]
Abstract
Class III Semaphorin (Sema) secreted ligands are known to repel neurites expressing Neuropilin (Nrp) and/or Plexin (Plxn) receptors. There is, however, a growing body of literature supporting that Sema signaling also has alternative roles in development such as synaptogenesis, boundary formation, and vasculogenesis. To evaluate these options during inner ear development, we used in situ hybridization or immunohistochemistry to map the expression of Sema3D, Sema3F, Nrp1, Nrp2, and PlxnA1 in the chicken (Gallus gallus) inner ear from embryonic day (E)5-E10. The resulting expression patterns in either the otic epithelium or its surrounding mesenchyme suggest that Sema signaling could be involved in each of the varied functions reported for other tissues. Sema3D expression flanking the sensory tissue in vestibular organs suggests that it may repel Nrp2- and PlxnA1-expressing neurites of the vestibular ganglion away from nonsensory epithelia, thus channeling them into the sensory domains at E5-E8. Expression of Sema signaling genes in the sensory hair cells of both the auditory and vestibular organs on E8-E10 may implicate Sema signaling in synaptogenesis. In the nonsensory regions of the cochlea, Sema3D in the future tegmentum vasculosum opposes Nrp1 and PlxnA1 in the future cuboidal cells; the abutment of ligand and receptors in adjacent domains may enforce or maintain the boundary between them. In the mesenchyme, Nrp1 colocalized with capillary-rich tissue. Sema3D immediately flanks this Nrp1-expressing tissue, suggesting a role in endothelial cell migration towards the inner ear. In summary, Sema signaling may play multiple roles in the developing inner ear.
Collapse
Affiliation(s)
- M. Katie Scott
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana 47907
| | - Jia Yue
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907
| | | | - Joo Won Lee
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907
| | - Donna M. Fekete
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana 47907
- Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907
| |
Collapse
|
25
|
Defourny J. Eph/ephrin signalling in the development and function of the mammalian cochlea. Dev Biol 2019; 449:35-40. [PMID: 30771305 DOI: 10.1016/j.ydbio.2019.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/10/2019] [Accepted: 02/11/2019] [Indexed: 12/12/2022]
Abstract
In mammals, the functional development of the cochlea requires the tight regulation of multiple molecules and signalling pathways including fibroblast growth factors, bone morphogenetic proteins, Wnt and Notch signalling pathways. Over the last decade, the Eph/ephrin system also emerged as a key player of the development and function of the mammalian cochlea. In this review, we discuss the recent advances on the role of Eph/ephrin signalling in patterning the cochlear sensory epithelium and the complex innervation of mechanosensory hair cells by spiral ganglion neurons. Finally, we address the issue of a syndromic form of hearing loss caused by a deficient member of the Eph/ephrin family.
Collapse
Affiliation(s)
- Jean Defourny
- GIGA-Neurosciences, Unit of Cell and Tissue Biology, University of Liège, C.H.U. B36, B-4000, Liège, Belgium.
| |
Collapse
|
26
|
Emerging Roles of Ephexins in Physiology and Disease. Cells 2019; 8:cells8020087. [PMID: 30682817 PMCID: PMC6406967 DOI: 10.3390/cells8020087] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 01/20/2019] [Accepted: 01/24/2019] [Indexed: 12/25/2022] Open
Abstract
Dbl (B-cell lymphoma)-related guanine nucleotide exchange factors (GEFs), the largest family of GEFs, are directly responsible for the activation of Rho family GTPases and essential for a number of cellular events such as proliferation, differentiation and movement. The members of the Ephexin (Eph-interacting exchange protein) family, a subgroup of Dbl GEFs, initially were named for their interaction with Eph receptors and sequence homology with Ephexin1. Although the first Ephexin was identified about two decades ago, their functions in physiological and pathological contexts and regulatory mechanisms remained elusive until recently. Ephexins are now considered as GEFs that can activate Rho GTPases such as RhoA, Rac, Cdc42, and RhoG. Moreover, Ephexins have been shown to have pivotal roles in neural development, tumorigenesis, and efferocytosis. In this review, we discuss the known and proposed functions of Ephexins in physiological and pathological contexts, as well as their regulatory mechanisms.
Collapse
|
27
|
Defourny J, Peuckert C, Kullander K, Malgrange B. EphA4-ADAM10 Interplay Patterns the Cochlear Sensory Epithelium through Local Disruption of Adherens Junctions. iScience 2018; 11:246-257. [PMID: 30639848 PMCID: PMC6327856 DOI: 10.1016/j.isci.2018.12.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 12/11/2022] Open
Abstract
The cochlear sensory epithelium contains a functionally important triangular fluid-filled space between adjacent pillar cells referred to as the tunnel of Corti. However, the molecular mechanisms leading to local cell-cell separation during development remain elusive. Here we show that EphA4 associates with ADAM10 to promote the destruction of E-cadherin-based adhesions between adjacent pillar cells. These cells fail to separate from each other, and E-cadherin abnormally persists at the pillar cell junction in EphA4 forward-signaling-deficient mice, as well as in the presence of ADAM10 inhibitor. Using immunolabeling and an in situ proximity ligation assay, we found that EphA4 forms a complex with E-cadherin and its sheddase ADAM10, which could be activated by ephrin-B2 across the pillar cell junction to trigger the cleavage of E-cadherin. Altogether, our findings provide a new molecular insight into the regulation of adherens junctions, which might be extended to a variety of physiological or pathological processes.
Collapse
Affiliation(s)
- Jean Defourny
- GIGA-Neurosciences, Unit of Cell and Tissue Biology, University of Liège, C.H.U. B36, 4000 Liège, Belgium; GIGA-Neurosciences, Developmental Neurobiology Unit, University of Liège, C.H.U. B36, 4000 Liège, Belgium
| | - Christiane Peuckert
- Department of Neuroscience, Uppsala University, Box 593, Uppsala 75124, Sweden
| | - Klas Kullander
- Department of Neuroscience, Uppsala University, Box 593, Uppsala 75124, Sweden
| | - Brigitte Malgrange
- GIGA-Neurosciences, Developmental Neurobiology Unit, University of Liège, C.H.U. B36, 4000 Liège, Belgium.
| |
Collapse
|
28
|
Petitpré C, Wu H, Sharma A, Tokarska A, Fontanet P, Wang Y, Helmbacher F, Yackle K, Silberberg G, Hadjab S, Lallemend F. Neuronal heterogeneity and stereotyped connectivity in the auditory afferent system. Nat Commun 2018; 9:3691. [PMID: 30209249 PMCID: PMC6135759 DOI: 10.1038/s41467-018-06033-3] [Citation(s) in RCA: 162] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 07/31/2018] [Indexed: 01/07/2023] Open
Abstract
Spiral ganglion (SG) neurons of the cochlea convey all auditory inputs to the brain, yet the cellular and molecular complexity necessary to decode the various acoustic features in the SG has remained unresolved. Using single-cell RNA sequencing, we identify four types of SG neurons, including three novel subclasses of type I neurons and the type II neurons, and provide a comprehensive genetic framework that define their potential synaptic communication patterns. The connectivity patterns of the three subclasses of type I neurons with inner hair cells and their electrophysiological profiles suggest that they represent the intensity-coding properties of auditory afferents. Moreover, neuron type specification is already established at birth, indicating a neuronal diversification process independent of neuronal activity. Thus, this work provides a transcriptional catalog of neuron types in the cochlea, which serves as a valuable resource for dissecting cell-type-specific functions of dedicated afferents in auditory perception and in hearing disorders.
Collapse
Affiliation(s)
- Charles Petitpré
- Department of Neuroscience, Karolinska Institutet, Biomedicum, Stockholm, 171 77, Sweden
| | - Haohao Wu
- Department of Neuroscience, Karolinska Institutet, Biomedicum, Stockholm, 171 77, Sweden
| | - Anil Sharma
- Department of Neuroscience, Karolinska Institutet, Biomedicum, Stockholm, 171 77, Sweden
| | - Anna Tokarska
- Department of Neuroscience, Karolinska Institutet, Biomedicum, Stockholm, 171 77, Sweden
| | - Paula Fontanet
- Department of Neuroscience, Karolinska Institutet, Biomedicum, Stockholm, 171 77, Sweden
| | - Yiqiao Wang
- Department of Neuroscience, Karolinska Institutet, Biomedicum, Stockholm, 171 77, Sweden
| | - Françoise Helmbacher
- Aix-Marseille Université, CNRS UMR7288, Institut de Biologie du Développement de Marseille (IBDM), 13009, Marseille, France
| | - Kevin Yackle
- Department of Physiology, University of California-San Francisco, San Francisco, CA, 94158, USA
| | - Gilad Silberberg
- Department of Neuroscience, Karolinska Institutet, Biomedicum, Stockholm, 171 77, Sweden
| | - Saida Hadjab
- Department of Neuroscience, Karolinska Institutet, Biomedicum, Stockholm, 171 77, Sweden
| | - François Lallemend
- Department of Neuroscience, Karolinska Institutet, Biomedicum, Stockholm, 171 77, Sweden.
| |
Collapse
|
29
|
Ghimire SR, Ratzan EM, Deans MR. A non-autonomous function of the core PCP protein VANGL2 directs peripheral axon turning in the developing cochlea. Development 2018; 145:dev.159012. [PMID: 29784671 DOI: 10.1242/dev.159012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 05/14/2018] [Indexed: 01/02/2023]
Abstract
The cochlea is innervated by neurons that relay sound information from hair cells to central auditory targets. A subset of these are the type II spiral ganglion neurons, which have nociceptive features and contribute to feedback circuits providing neuroprotection in extreme noise. Type II neurons make a distinctive 90° turn towards the cochlear base to synapse with 10-15 outer hair cells. We demonstrate that this axon turning event requires planar cell polarity (PCP) signaling and is disrupted in Vangl2 and Celsr1 knockout mice, and that VANGL2 acts non-autonomously from the cochlea to direct turning. Moreover, VANGL2 is asymmetrically distributed at intercellular junctions between cochlear supporting cells, and in a pattern that could allow it to act directly as an axon guidance cue. Together, these data reveal a non-autonomous function for PCP signaling during axon guidance occurring in the tissue that is innervated, rather than the navigating growth cone.
Collapse
Affiliation(s)
- Satish R Ghimire
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Evan M Ratzan
- Interdepartmental Program in Neuroscience, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Michael R Deans
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA .,Interdepartmental Program in Neuroscience, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.,Department of Surgery, Division of Otolaryngology, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| |
Collapse
|
30
|
Ephexin1 Is Required for Eph-Mediated Limb Trajectory of Spinal Motor Axons. J Neurosci 2018; 38:2043-2056. [PMID: 29363583 DOI: 10.1523/jneurosci.2257-17.2018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 01/10/2018] [Accepted: 01/15/2018] [Indexed: 11/21/2022] Open
Abstract
The precise assembly of a functional nervous system relies on the guided migration of axonal growth cones, which is made possible by signals transmitted to the cytoskeleton by cell surface-expressed guidance receptors. We investigated the function of ephexin1, a Rho guanine nucleotide exchange factor, as an essential growth-cone guidance intermediary in the context of spinal lateral motor column (LMC) motor axon trajectory selection in the limb mesenchyme. Using in situ mRNA detection, we first show that ephexin1 is expressed in LMC neurons of chick and mouse embryos at the time of spinal motor axon extension into the limb. Ephexin1 loss of function and gain of function using in ovo electroporation in chick LMC neurons, of either sex, perturbed LMC axon trajectory selection, demonstrating an essential role of ephexin1 in motor axon guidance. In addition, ephexin1 loss in mice of either sex led to LMC axon trajectory selection errors. We also show that ephexin1 knockdown attenuates the growth preference of LMC neurites against ephrins in vitro and Eph receptor-mediated retargeting of LMC axons in vivo, suggesting that ephexin1 is required in Eph-mediated LMC motor axon guidance. Finally, both ephexin1 knockdown and ectopic expression of nonphosphorylatable ephexin1 mutant attenuated the retargeting of LMC axons caused by Src overexpression, implicating ephexin1 as an Src target in Eph signal relay in this context. In summary, our findings demonstrate that ephexin1 is essential for motor axon guidance and suggest an important role in relaying ephrin:Eph signals that mediate motor axon trajectory selection.SIGNIFICANCE STATEMENT The proper development of functioning neural circuits requires precise nerve connections among neurons or between neurons and their muscle targets. The Eph tyrosine kinase receptors expressed in neurons are important in many contexts during neural-circuit formation, such as axon outgrowth, axon guidance, and synaptic formation, and have been suggested to be involved in neurodegenerative disorders, including amyotrophic lateral sclerosis and Alzheimer's disease. To dissect the mechanism of Eph signal relay, we studied ephexin1 gain of function and loss of function and found ephexin1 essential for the development of limb nerves toward their muscle targets, concluding that it functions as an intermediary to relay Eph signaling in this context. Our work could thus shed new light on the molecular mechanisms controlling neuromuscular connectivity during embryonic development.
Collapse
|
31
|
Tuft BW, Xu L, Leigh B, Lee D, Guymon CA, Hansen MR. Photopolymerized micropatterns with high feature frequencies overcome chemorepulsive borders to direct neurite growth. J Tissue Eng Regen Med 2017; 12:e1392-e1403. [PMID: 28753740 DOI: 10.1002/term.2527] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 07/03/2017] [Accepted: 07/24/2017] [Indexed: 01/04/2023]
Abstract
Developing and regenerating neurites respond to a variety of biophysical and biochemical cues in their micro-environment to reach target cells and establish appropriate synapses. Defining the hierarchal relationship of both types of cues to direct neurite growth carries broad significance for neural development, regeneration, and, in particular, engineering of neural prostheses that improve tissue integration with native neural networks. In this work, chemorepulsive biochemical borders are established on substrates with a range of surface microfeatures to determine the potential of physical cues to overcome conflicting biochemical cues. Physical micropatterns are fabricated using photomasking techniques to spatially control photoinitiation events of the polymerization. Temporal control of the reaction allows for generation of microfeatures with the same amplitude across a range of feature frequencies or periodicities. The micropatterned substrates are then modified with repulsive chemical borders between laminin and either EphA4-Fc or tenascin C that compete with the surface microfeatures to direct neurite growth. Behaviour of neurites from spiral ganglion and trigeminal neurons is characterized at biochemical borders as cross, turn, stop, or repel events. Both the chemical borders and physical patterns significantly influence neurite pathfinding. On unpatterned surfaces, most neurites that originate on laminin are deterred by the border with tenascin C or EphA4-Fc. Importantly, substrates with frequent micropattern features overcome the influence of the chemorepulsive border to dominate neurite trajectory. Designing prosthesis interfaces with appropriate surface features may allow for spatially organized neurite outgrowth in vivo even in the presence of conflicting biochemical cues in native target tissues.
Collapse
Affiliation(s)
- Bradley W Tuft
- Department of Chemical and Biochemical Engineering, University of Iowa, Iowa City, IA, USA
| | - Linjing Xu
- Department of Otolaryngology - Head and Neck Surgery, University of Iowa, Iowa City, IA, USA
| | - Braden Leigh
- Department of Chemical and Biochemical Engineering, University of Iowa, Iowa City, IA, USA
| | - Daniel Lee
- Department of Otolaryngology - Head and Neck Surgery, University of Iowa, Iowa City, IA, USA
| | - C Allan Guymon
- Department of Chemical and Biochemical Engineering, University of Iowa, Iowa City, IA, USA
| | - Marlan R Hansen
- Department of Otolaryngology - Head and Neck Surgery, University of Iowa, Iowa City, IA, USA.,Department of Neurosurgery, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
32
|
Ingham NJ, Steel KP, Drescher U. On the role of ephrinA2 in auditory function. Hear Res 2017; 350:11-16. [DOI: 10.1016/j.heares.2017.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/24/2017] [Accepted: 04/03/2017] [Indexed: 11/24/2022]
|
33
|
Cheng C, Guo L, Lu L, Xu X, Zhang S, Gao J, Waqas M, Zhu C, Chen Y, Zhang X, Xuan C, Gao X, Tang M, Chen F, Shi H, Li H, Chai R. Characterization of the Transcriptomes of Lgr5+ Hair Cell Progenitors and Lgr5- Supporting Cells in the Mouse Cochlea. Front Mol Neurosci 2017; 10:122. [PMID: 28491023 PMCID: PMC5405134 DOI: 10.3389/fnmol.2017.00122] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 04/11/2017] [Indexed: 12/27/2022] Open
Abstract
Cochlear supporting cells (SCs) have been shown to be a promising resource for hair cell (HC) regeneration in the neonatal mouse cochlea. Previous studies have reported that Lgr5+ SCs can regenerate HCs both in vitro and in vivo and thus are considered to be inner ear progenitor cells. Lgr5+ progenitors are able to regenerate more HCs than Lgr5- SCs, and it is important to understand the mechanism behind the proliferation and HC regeneration of these progenitors. Here, we isolated Lgr5+ progenitors and Lgr5- SCs from Lgr5-EGFP-CreERT2/Sox2-CreERT2/Rosa26-tdTomato mice via flow cytometry. As expected, we found that Lgr5+ progenitors had significantly higher proliferation and HC regeneration ability than Lgr5- SCs. Next, we performed RNA-Seq to determine the gene expression profiles of Lgr5+ progenitors and Lgr5- SCs. We analyzed the genes that were enriched and differentially expressed in Lgr5+ progenitors and Lgr5- SCs, and we found 8 cell cycle genes, 9 transcription factors, and 24 cell signaling pathway genes that were uniquely expressed in one population but not the other. Last, we made a protein–protein interaction network to further analyze the role of these differentially expressed genes. In conclusion, we present a set of genes that might regulate the proliferation and HC regeneration ability of Lgr5+ progenitors, and these might serve as potential new therapeutic targets for HC regeneration.
Collapse
Affiliation(s)
- Cheng Cheng
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China.,Research Institute of OtolaryngologyNanjing, China.,Co-innovation Center of Neuroregeneration, Nantong UniversityNantong, China
| | - Luo Guo
- Department of Otorhinolaryngology, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China.,Key Laboratory of Hearing Medicine of National Health and Family Planning CommissionShanghai, China
| | - Ling Lu
- Department of Otolaryngology-Head and Neck Surgery, Nanjing Drum Tower Hospital, Nanjing University Medical SchoolNanjing, China.,Department of Otolaryngology-Head and Neck Surgery, Drum Tower Clinical Medical College of Nanjing Medical UniversityNanjing, China
| | - Xiaochen Xu
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China
| | - ShaSha Zhang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China
| | - Junyan Gao
- Health Management and Policy, College of Public Health, Saint Louis University, St. LouisMO, USA
| | - Muhammad Waqas
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China.,Department of Biotechnology, Federal Urdu University of Arts, Science and TechnologyGulshan-e-Iqbal, Pakistan
| | - Chengwen Zhu
- Department of Otolaryngology-Head and Neck Surgery, Nanjing Drum Tower Hospital, Nanjing University Medical SchoolNanjing, China
| | - Yan Chen
- Department of Otorhinolaryngology, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China.,Key Laboratory of Hearing Medicine of National Health and Family Planning CommissionShanghai, China
| | - Xiaoli Zhang
- Department of Otolaryngology-Head and Neck Surgery, Nanjing Drum Tower Hospital, Nanjing University Medical SchoolNanjing, China
| | - Chuanying Xuan
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China
| | - Xia Gao
- Department of Otolaryngology-Head and Neck Surgery, Nanjing Drum Tower Hospital, Nanjing University Medical SchoolNanjing, China
| | - Mingliang Tang
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China
| | - Fangyi Chen
- Department of Biomedical Engineering, Southern University of Science and TechnologyShenzhen, China
| | - Haibo Shi
- Department of Otorhinolaryngology Head and Neck Surgery, The Sixth People's Hospital, Shanghai Jiao Tong UniversityShanghai, China
| | - Huawei Li
- Department of Otorhinolaryngology, Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan UniversityShanghai, China.,Key Laboratory of Hearing Medicine of National Health and Family Planning CommissionShanghai, China.,Institutes of Biomedical Sciences, Fudan UniversityShanghai, China.,Shanghai Engineering Research Centre of Cochlear ImplantShanghai, China
| | - Renjie Chai
- Key Laboratory for Developmental Genes and Human Disease, Ministry of Education, Institute of Life Sciences, Southeast UniversityNanjing, China.,Research Institute of OtolaryngologyNanjing, China.,Co-innovation Center of Neuroregeneration, Nantong UniversityNantong, China
| |
Collapse
|
34
|
Zhang KD, Coate TM. Recent advances in the development and function of type II spiral ganglion neurons in the mammalian inner ear. Semin Cell Dev Biol 2016; 65:80-87. [PMID: 27760385 DOI: 10.1016/j.semcdb.2016.09.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 08/12/2016] [Accepted: 09/25/2016] [Indexed: 01/17/2023]
Abstract
In hearing, mechanically sensitive hair cells (HCs) in the cochlea release glutamate onto spiral ganglion neurons (SGNs) to relay auditory information to the central nervous system (CNS). There are two main SGN subtypes, which differ in morphology, number, synaptic targets, innervation patterns and firing properties. About 90-95% of SGNs are the type I SGNs, which make a single bouton connection with inner hair cells (IHCs) and have been well described in the canonical auditory pathway for sound detection. However, less attention has been given to the type II SGNs, which exclusively innervate outer hair cells (OHCs). In this review, we emphasize recent advances in the molecular mechanisms that control how type II SGNs develop and form connections with OHCs, and exciting new insights into the function of type II SGNs.
Collapse
Affiliation(s)
- Kaidi D Zhang
- Department of Biology, Georgetown University, Washington, DC, USA.
| | - Thomas M Coate
- Department of Biology, Georgetown University, Washington, DC, USA
| |
Collapse
|
35
|
Saito Y, Miranda-Rottmann S, Ruggiu M, Park CY, Fak JJ, Zhong R, Duncan JS, Fabella BA, Junge HJ, Chen Z, Araya R, Fritzsch B, Hudspeth AJ, Darnell RB. NOVA2-mediated RNA regulation is required for axonal pathfinding during development. eLife 2016; 5. [PMID: 27223325 PMCID: PMC4930328 DOI: 10.7554/elife.14371] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 05/23/2016] [Indexed: 01/13/2023] Open
Abstract
The neuron specific RNA-binding proteins NOVA1 and NOVA2 are highly homologous alternative splicing regulators. NOVA proteins regulate at least 700 alternative splicing events in vivo, yet relatively little is known about the biologic consequences of NOVA action and in particular about functional differences between NOVA1 and NOVA2. Transcriptome-wide searches for isoform-specific functions, using NOVA1 and NOVA2 specific HITS-CLIP and RNA-seq data from mouse cortex lacking either NOVA isoform, reveals that NOVA2 uniquely regulates alternative splicing events of a series of axon guidance related genes during cortical development. Corresponding axonal pathfinding defects were specific to NOVA2 deficiency: Nova2-/- but not Nova1-/- mice had agenesis of the corpus callosum, and axonal outgrowth defects specific to ventral motoneuron axons and efferent innervation of the cochlea. Thus we have discovered that NOVA2 uniquely regulates alternative splicing of a coordinate set of transcripts encoding key components in cortical, brainstem and spinal axon guidance/outgrowth pathways during neural differentiation, with severe functional consequences in vivo.
Collapse
Affiliation(s)
- Yuhki Saito
- Laboratory of Molecular Neuro-Oncology, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Soledad Miranda-Rottmann
- Laboratory of Molecular Neuro-Oncology, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Matteo Ruggiu
- Laboratory of Molecular Neuro-Oncology, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | | | - John J Fak
- Laboratory of Molecular Neuro-Oncology, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Ru Zhong
- Laboratory of Molecular Neuro-Oncology, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Jeremy S Duncan
- Department of Biology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, United States
| | - Brian A Fabella
- Laboratory of Sensory Neuroscience, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Harald J Junge
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Boulder, United States
| | - Zhe Chen
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Boulder, United States
| | - Roberto Araya
- Department of Neurosciences, Faculty of Medicine, University of Montreal, Montreal, Canada
| | - Bernd Fritzsch
- Department of Biology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, United States
| | - A J Hudspeth
- Laboratory of Sensory Neuroscience, Howard Hughes Medical Institute, The Rockefeller University, New York, United States
| | - Robert B Darnell
- Laboratory of Molecular Neuro-Oncology, Howard Hughes Medical Institute, The Rockefeller University, New York, United States.,New York Genome Center, New York, United States
| |
Collapse
|
36
|
Abstract
UNLABELLED A hallmark of the nervous system is the presence of precise patterns of connections between different types of neurons. Many mechanisms can be used to establish specificity, including homophilic adhesion and synaptic refinement, but the range of strategies used across the nervous system remains unclear. To broaden the understanding of how neurons find their targets, we studied the developing murine cochlea, where two classes of spiral ganglion neurons (SGNs), type I and type II, navigate together to the sensory epithelium and then diverge to contact inner hair cells (IHCs) or outer hair cells (OHCs), respectively. Neurons with type I and type II morphologies are apparent before birth, suggesting that target selection might be accomplished by excluding type I processes from the OHC region. However, because type I processes appear to overshoot into type II territory postnatally, specificity may also depend on elimination of inappropriate synapses. To resolve these differences, we analyzed the morphology and dynamic behaviors of individual fibers and their branches as they interact with potential partners. We found that SGN processes continue to be segregated anatomically in the postnatal cochlea. Although type I-like fibers branched locally, few branches contacted OHCs, arguing against synaptic elimination. Instead, time-lapse imaging studies suggest a prominent role for retraction, first positioning processes to the appropriate region and then corralling branches during a subsequent period of exuberant growth and refinement. Thus, sequential stages of retraction can help to achieve target specificity, adding to the list of mechanisms available for sculpting neural circuits. SIGNIFICANCE STATEMENT During development, different types of neurons must form connections with specific synaptic targets, thereby creating the precise wiring diagram necessary for adult function. Although studies have revealed multiple mechanisms for target selection, we still know little about how different strategies are used to produce each circuit's unique pattern of connectivity. Here we combined neurite-tracing and time-lapse imaging to define the events that lead to the simple binary wiring specificity of the cochlea. A better understanding of how the cochlea is innervated will broaden our knowledge of target selection across the nervous system, offer new insights into the developmental origins of deafness, and guide efforts to restore connectivity in the damaged cochlea.
Collapse
|
37
|
Dcc Mediates Functional Assembly of Peripheral Auditory Circuits. Sci Rep 2016; 6:23799. [PMID: 27040640 PMCID: PMC4819185 DOI: 10.1038/srep23799] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 03/11/2016] [Indexed: 01/12/2023] Open
Abstract
Proper structural organization of spiral ganglion (SG) innervation is crucial for normal hearing function. However, molecular mechanisms underlying the developmental formation of this precise organization remain not well understood. Here, we report in the developing mouse cochlea that deleted in colorectal cancer (Dcc) contributes to the proper organization of spiral ganglion neurons (SGNs) within the Rosenthal's canal and of SGN projections toward both the peripheral and central auditory targets. In Dcc mutant embryos, mispositioning of SGNs occurred along the peripheral auditory pathway with misrouted afferent fibers and reduced synaptic contacts with hair cells. The central auditory pathway simultaneously exhibited similar defective phenotypes as in the periphery with abnormal exit of SGNs from the Rosenthal's canal towards central nuclei. Furthermore, the axons of SGNs ascending into the cochlear nucleus had disrupted bifurcation patterns. Thus, Dcc is necessary for establishing the proper spatial organization of SGNs and their fibers in both peripheral and central auditory pathways, through controlling axon targeting and cell migration. Our results suggest that Dcc plays an important role in the developmental formation of peripheral and central auditory circuits, and its mutation may contribute to sensorineural hearing loss.
Collapse
|
38
|
Treffy RW, Collins D, Hoshino N, Ton S, Katsevman GA, Oleksiak M, Runge EM, Cho D, Russo M, Spec A, Gomulka J, Henkemeyer M, Rochlin MW. Ephrin-B/EphB Signaling Is Required for Normal Innervation of Lingual Gustatory Papillae. Dev Neurosci 2016; 38:124-38. [PMID: 27035151 PMCID: PMC4927353 DOI: 10.1159/000444748] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 02/17/2016] [Indexed: 02/02/2023] Open
Abstract
The innervation of taste buds is an excellent model system for studying the guidance of axons during targeting because of their discrete nature and the high fidelity of innervation. The pregustatory epithelium of fungiform papillae is known to secrete diffusible axon guidance cues such as BDNF and Sema3A that attract and repel, respectively, geniculate ganglion axons during targeting, but diffusible factors alone are unlikely to explain how taste axon terminals are restricted to their territories within the taste bud. Nondiffusible cell surface proteins such as Ephs and ephrins can act as receptors and/or ligands for one another and are known to control axon terminal positioning in several parts of the nervous system, but they have not been studied in the gustatory system. We report that ephrin-B2 linked β-galactosidase staining and immunostaining was present along the dorsal epithelium of the mouse tongue as early as embryonic day 15.5 (E15.5), but was not detected at E14.5, when axons first enter the epithelium. Ephrin-B1 immunolabeling was barely detected in the epithelium and found at a somewhat higher concentration in the mesenchyme subjacent to the epithelium. EphB1 and EphB2 were detected in lingual sensory afferents in vivo and geniculate neurites in vitro. Ephrin-B1 and ephrin-B2 were similarly effective in repelling or suppressing outgrowth by geniculate neurites in vitro. These in vitro effects were independent of the neurotrophin used to promote outgrowth, but were reduced by elevated levels of laminin. In vivo, mice null for EphB1 and EphB2 exhibited decreased gustatory innervation of fungiform papillae. These data provide evidence that ephrin-B forward signaling is necessary for normal gustatory innervation of the mammalian tongue.
Collapse
|
39
|
Wallace MM, Harris JA, Brubaker DQ, Klotz CA, Gabriele ML. Graded and discontinuous EphA-ephrinB expression patterns in the developing auditory brainstem. Hear Res 2016; 335:64-75. [PMID: 26906676 DOI: 10.1016/j.heares.2016.02.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Revised: 02/02/2016] [Accepted: 02/18/2016] [Indexed: 01/06/2023]
Abstract
Eph-ephrin interactions guide topographic mapping and pattern formation in a variety of systems. In contrast to other sensory pathways, their precise role in the assembly of central auditory circuits remains poorly understood. The auditory midbrain, or inferior colliculus (IC) is an intriguing structure for exploring guidance of patterned projections as adjacent subdivisions exhibit distinct organizational features. The central nucleus of the IC (CNIC) and deep aspects of its neighboring lateral cortex (LCIC, Layer 3) are tonotopically-organized and receive layered inputs from primarily downstream auditory sources. While less is known about more superficial aspects of the LCIC, its inputs are multimodal, lack a clear tonotopic order, and appear discontinuous, terminating in modular, patch/matrix-like distributions. Here we utilize X-Gal staining approaches in lacZ mutant mice (ephrin-B2, -B3, and EphA4) to reveal EphA-ephrinB expression patterns in the nascent IC during the period of projection shaping that precedes hearing onset. We also report early postnatal protein expression in the cochlear nuclei, the superior olivary complex, the nuclei of the lateral lemniscus, and relevant midline structures. Continuous ephrin-B2 and EphA4 expression gradients exist along frequency axes of the CNIC and LCIC Layer 3. In contrast, more superficial LCIC localization is not graded, but confined to a series of discrete ephrin-B2 and EphA4-positive Layer 2 modules. While heavily expressed in the midline, much of the auditory brainstem is devoid of ephrin-B3, including the CNIC, LCIC Layer 2 modular fields, the dorsal nucleus of the lateral lemniscus (DNLL), as well as much of the superior olivary complex and cochlear nuclei. Ephrin-B3 LCIC expression appears complementary to that of ephrin-B2 and EphA4, with protein most concentrated in presumptive extramodular zones. Described tonotopic gradients and seemingly complementary modular/extramodular patterns suggest Eph-ephrin guidance in establishing juxtaposed continuous and discrete neural maps in the developing IC prior to experience.
Collapse
Affiliation(s)
- Matthew M Wallace
- James Madison University, Department of Biology, Harrisonburg, VA 22807, USA
| | - J Aaron Harris
- James Madison University, Department of Biology, Harrisonburg, VA 22807, USA
| | - Donald Q Brubaker
- James Madison University, Department of Biology, Harrisonburg, VA 22807, USA
| | - Caitlyn A Klotz
- James Madison University, Department of Biology, Harrisonburg, VA 22807, USA
| | - Mark L Gabriele
- James Madison University, Department of Biology, Harrisonburg, VA 22807, USA.
| |
Collapse
|
40
|
Abstract
The identification of transcriptional differences has served as an important starting point in understanding the molecular mechanisms behind biological processes and systems. The developmental biology of the inner ear, the biology of hearing and of course the pathology of deafness are all processes that warrant a molecular description if we are to improve human health. To this end, technological innovation has meant that larger scale analysis of gene transcription has been possible for a number of years now, extending our molecular analysis of genes to beyond those that are currently in vogue for a given system. In this review, some of the contributions gene profiling has made to understanding developmental, pathological and physiological processes in the inner ear are highlighted.
Collapse
Affiliation(s)
- Thomas Schimmang
- Instituto de Biología y Genética MolecularUniversidad de Valladolid y Consejo Superior de Investigaciones CientíficasValladolidSpain
| | - Mark Maconochie
- School of Biological and Chemical SciencesQueen Mary University of LondonLondonUK
| |
Collapse
|
41
|
Goodrich LV. Early Development of the Spiral Ganglion. THE PRIMARY AUDITORY NEURONS OF THE MAMMALIAN COCHLEA 2016. [DOI: 10.1007/978-1-4939-3031-9_2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
42
|
Abdul-Latif ML, Salazar JAA, Marshak S, Dinh ML, Cramer KS. Ephrin-A2 and ephrin-A5 guide contralateral targeting but not topographic mapping of ventral cochlear nucleus axons. Neural Dev 2015; 10:27. [PMID: 26666565 PMCID: PMC4678660 DOI: 10.1186/s13064-015-0054-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 12/10/2015] [Indexed: 11/23/2022] Open
Abstract
Background In the auditory brainstem, ventral cochlear nucleus (VCN) axons project to the contralateral, but not ipsilateral, medial nucleus of trapezoid body (MNTB), terminating in the calyx of Held. Dorsal VCN neurons, representing high frequencies, synapse with medial MNTB neurons, while low frequency-coding ventral VCN neurons synapse with lateral MNTB neurons, reflecting tonotopic organization. The mechanisms that ensure strictly contralateral targeting and topographic ordering are incompletely understood. Here we examined the roles of ephrin-A signaling in both types of targeting. Results Ephrin-A2 and ephrin-A5 are expressed in VCN cells during late embryonic and early postnatal development. At these ages ephrin-A2 is expressed in axons surrounding MNTB and ephrin-A5 is expressed in MNTB principal neurons. Ephrin-A2/A5 double knockout mice displayed axon targeting errors in which VCN axons project to MNTB on both sides of the brainstem, where they terminate in calyceal endings. Ephrin-A2 and ephrin-A5 single knockout mice showed a similar phenotype. In contrast to effects on contralateral targeting, ephrin-A2/A5 double knockout mice showed no defects in formation of tonotopically ordered projections from VCN to MNTB. Conclusions These findings demonstrate that distinct mechanisms regulate targeting of VCN axons to the contralateral MNTB and targeting to appropriate tonotopic locations. Ephrin-A signaling plays a similar role to ephrin-B signaling in the VCN-MNTB pathway, where both classes normally prevent formation of calyceal projections to ipsilateral MNTB. These classes may rely in part on common signaling pathways.
Collapse
Affiliation(s)
- Mariam L Abdul-Latif
- Division of Neonatology, Department of Pediatrics, University of California, Irvine, 101 The City Drive, Orange, CA, 92868-3298, USA.
| | - Jesus A Ayala Salazar
- Department of Neurobiology and Behavior, University of California, Irvine, 2205 McGaugh Hall, Irvine, CA, 92697-4550, USA.
| | - Sonya Marshak
- Department of Neurobiology and Behavior, University of California, Irvine, 2205 McGaugh Hall, Irvine, CA, 92697-4550, USA.
| | - Minhan L Dinh
- Department of Neurobiology and Behavior, University of California, Irvine, 2205 McGaugh Hall, Irvine, CA, 92697-4550, USA.
| | - Karina S Cramer
- Department of Neurobiology and Behavior, University of California, Irvine, 2205 McGaugh Hall, Irvine, CA, 92697-4550, USA.
| |
Collapse
|
43
|
Cochlear afferent innervation development. Hear Res 2015; 330:157-69. [DOI: 10.1016/j.heares.2015.07.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 06/02/2015] [Accepted: 07/21/2015] [Indexed: 01/11/2023]
|
44
|
Sundaresan S, Balasubbu S, Mustapha M. Thyroid hormone is required for the pruning of afferent type II spiral ganglion neurons in the mouse cochlea. Neuroscience 2015; 312:165-78. [PMID: 26592716 DOI: 10.1016/j.neuroscience.2015.11.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 11/09/2015] [Accepted: 11/10/2015] [Indexed: 12/13/2022]
Abstract
Afferent connections to the sensory inner (IHCs) and outer hair cells (OHCs) in the cochlea refine and functionally mature during the thyroid hormone (TH)-critical period of inner ear development that occurs perinatally in rodents. In this study, we investigated the effects of hypothyroidism on afferent type II innervation to outer hair cells using the Snell dwarf mouse (Pit1(dw)). Using a transgenic approach to specifically label type II spiral ganglion neurons (SGNs), we found that lack of TH causes persistence of excess type II SGN connections to the OHCs, as well as continued expression of the hair cell functional marker, otoferlin (OTOF), in the OHCs beyond the maturation period. We also observed a concurrent delay in efferent attachment to the OHCs. Supplementing with TH during the early postnatal period from postnatal day (P) 3 to P4 reversed the defect in type II SGN pruning but did not alter OTOF expression. Our results show that hypothyroidism causes a defect in the large-scale pruning of afferent type II SGNs in the cochlea, and a delay in efferent attachment and the maturation of OTOF expression. Our data suggest that the state of maturation of hair cells, as determined by OTOF expression, may not regulate the pruning of their afferent innervation.
Collapse
Affiliation(s)
- S Sundaresan
- Department of Otolaryngology-Head & Neck Surgery, 300 Pasteur Drive, Stanford University, Stanford, CA 94035, United States
| | - S Balasubbu
- Department of Otolaryngology-Head & Neck Surgery, 300 Pasteur Drive, Stanford University, Stanford, CA 94035, United States
| | - M Mustapha
- Department of Otolaryngology-Head & Neck Surgery, 300 Pasteur Drive, Stanford University, Stanford, CA 94035, United States.
| |
Collapse
|
45
|
Terriente J, Pujades C. Cell segregation in the vertebrate hindbrain: a matter of boundaries. Cell Mol Life Sci 2015; 72:3721-30. [PMID: 26089248 PMCID: PMC11113478 DOI: 10.1007/s00018-015-1953-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 05/06/2015] [Accepted: 06/08/2015] [Indexed: 02/07/2023]
Abstract
Segregating cells into compartments during embryonic development is essential for growth and pattern formation. In the developing hindbrain, boundaries separate molecularly, physically and neuroanatomically distinct segments called rhombomeres. After rhombomeric cells have acquired their identity, interhombomeric boundaries restrict cell intermingling between adjacent rhombomeres and act as signaling centers to pattern the surrounding tissue. Several works have stressed the relevance of Eph/ephrin signaling in rhombomeric cell sorting. Recent data have unveiled the role of this pathway in the assembly of actomyosin cables as an important mechanism for keeping cells from different rhombomeres segregated. In this Review, we will provide a short summary of recent evidences gathered in different systems suggesting that physical actomyosin barriers can be a general mechanism for tissue separation. We will discuss current evidences supporting a model where cell-cell signaling pathways, such as Eph/ephrin, govern compartmental cell sorting through modulation of the actomyosin cytoskeleton and cell adhesive properties to prevent cell intermingling.
Collapse
Affiliation(s)
- Javier Terriente
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, PRBB, Dr Aiguader 88, 08003, Barcelona, Spain.
| | - Cristina Pujades
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, PRBB, Dr Aiguader 88, 08003, Barcelona, Spain.
| |
Collapse
|
46
|
Coate TM, Spita NA, Zhang KD, Isgrig KT, Kelley MW. Neuropilin-2/Semaphorin-3F-mediated repulsion promotes inner hair cell innervation by spiral ganglion neurons. eLife 2015; 4. [PMID: 26302206 PMCID: PMC4566076 DOI: 10.7554/elife.07830] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 08/22/2015] [Indexed: 12/18/2022] Open
Abstract
Auditory function is dependent on the formation of specific innervation patterns between mechanosensory hair cells (HCs) and afferent spiral ganglion neurons (SGNs). In particular, type I SGNs must precisely connect with inner HCs (IHCs) while avoiding connections with nearby outer HCs (OHCs). The factors that mediate these patterning events are largely unknown. Using sparse-labeling and time-lapse imaging, we visualized for the first time the behaviors of developing SGNs including active retraction of processes from OHCs, suggesting that some type I SGNs contact OHCs before forming synapses with IHCs. In addition, we demonstrate that expression of Semaphorin-3F in the OHC region inhibits type I SGN process extension by activating Neuropilin-2 receptors expressed on SGNs. These results suggest a model in which cochlear innervation patterns by type I SGNs are determined, at least in part, through a Semaphorin-3F-mediated inhibitory signal that impedes processes from extending beyond the IHC region. DOI:http://dx.doi.org/10.7554/eLife.07830.001 The process of hearing begins when sound waves enter the outer ear, causing the eardrum to vibrate. The three small bones of the middle ear pass these vibrations on to the cochlea, a fluid-filled structure shaped like a spiral. Tiny hair cells inside the cochlea move in response to the vibrations and convert them into electrical signals, which are transmitted by cells called spiral ganglion neurons (SGNs) to the brain. Hair cells can be divided into ‘inner’ and ‘outer’ hair cells. Inner hair cells transmit most of the information about a sound to the brain, via connections with type I SGNs. Outer hair cells are thought to amplify sound and connect to type II SGNs. How the type I and II SGNs connect to the correct type of hair cell as the ear develops is not well understood, despite these connections being essential for hearing. Coate et al. have now used time-lapse imaging and fixed specimens to follow individually labeled SGNs as they establish these connections within the cochlea of a mouse embryo. Although the type I SGNs ultimately formed connections with inner hair cells, many of them made contact with outer hair cells first. These contacts were short-lived thanks to a protein found near the outer hair cells, named Semaphorin-3F. This protein repels the type I SGNs by activating a receptor on their surface called Neuropilin-2, and so directs the type I SGNs towards the inner hair cells. One of the mysteries that remains to be solved is how type II SGNs are ‘permitted’ to extend into the outer hair cell region, even though they are also confronted by Semaphorin-3F. In addition, it will also be important to determine how SGNs adapt to cues from different Semaphorins from different parts of the cochlea as they navigate into different hair cell regions. DOI:http://dx.doi.org/10.7554/eLife.07830.002
Collapse
Affiliation(s)
- Thomas M Coate
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, Bethesda, United States
| | - Nathalie A Spita
- Department of Biology, Georgetown University, Washington, United States
| | - Kaidi D Zhang
- Department of Biology, Georgetown University, Washington, United States
| | - Kevin T Isgrig
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, Bethesda, United States
| | - Matthew W Kelley
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, Bethesda, United States
| |
Collapse
|
47
|
Kim YJ, Ibrahim LA, Wang SZ, Yuan W, Evgrafov OV, Knowles JA, Wang K, Tao HW, Zhang LI. EphA7 regulates spiral ganglion innervation of cochlear hair cells. Dev Neurobiol 2015; 76:452-69. [PMID: 26178595 DOI: 10.1002/dneu.22326] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 06/10/2015] [Accepted: 07/14/2015] [Indexed: 01/11/2023]
Abstract
During the development of periphery auditory circuitry, spiral ganglion neurons (SGNs) form a spatially precise pattern of innervation of cochlear hair cells (HCs), which is an essential structural foundation for central auditory processing. However, molecular mechanisms underlying the developmental formation of this precise innervation pattern remain not well understood. Here, we specifically examined the involvement of Eph family members in cochlear development. By performing RNA-sequencing for different types of cochlear cell, in situ hybridization, and immunohistochemistry, we found that EphA7 was strongly expressed in a large subset of SGNs. In EphA7 deletion mice, there was a reduction in the number of inner radial bundles originating from SGNs and projecting to HCs as well as in the number of ribbon synapses on inner hair cells (IHCs), as compared with wild-type or heterozygous mutant mice, attributable to fewer type I afferent fibers. The overall activity of the auditory nerve in EphA7 deletion mice was also reduced, although there was no significant change in the hearing intensity threshold. In vitro analysis further suggested that the reduced innervation of HCs by SGNs could be attributed to a role of EphA7 in regulating outgrowth of SGN neurites as knocking down EphA7 in SGNs resulted in diminished SGN fibers. In addition, suppressing the activity of ERK1/2, a potential downstream target of EphA7 signaling, either with specific inhibitors in cultured explants or by knocking out Prkg1, also resulted in reduced SGN fibers. Together, our results suggest that EphA7 plays an important role in the developmental formation of cochlear innervation pattern through controlling SGN fiber ontogeny. Such regulation may contribute to the salience level of auditory signals presented to the central auditory system.
Collapse
Affiliation(s)
- Young J Kim
- Zilkha Neurogenetic Institute, Keck School of Medicine, University Of Southern California, Los Angeles, California, 90033.,Neuroscience Graduate Program, University Of Southern California, Los Angeles, California
| | - Leena A Ibrahim
- Zilkha Neurogenetic Institute, Keck School of Medicine, University Of Southern California, Los Angeles, California, 90033.,Neuroscience Graduate Program, University Of Southern California, Los Angeles, California
| | - Sheng-Zhi Wang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University Of Southern California, Los Angeles, California, 90033
| | - Wei Yuan
- Department of Otolaryngology of Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Oleg V Evgrafov
- Zilkha Neurogenetic Institute, Keck School of Medicine, University Of Southern California, Los Angeles, California, 90033.,Department of Psychiatry, Keck School Of Medicine, University Of Southern California, Los Angeles, California
| | - James A Knowles
- Zilkha Neurogenetic Institute, Keck School of Medicine, University Of Southern California, Los Angeles, California, 90033.,Department of Psychiatry, Keck School Of Medicine, University Of Southern California, Los Angeles, California
| | - Kai Wang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University Of Southern California, Los Angeles, California, 90033.,Department of Psychiatry, Keck School Of Medicine, University Of Southern California, Los Angeles, California
| | - Huizhong W Tao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University Of Southern California, Los Angeles, California, 90033.,Department of Cell And Neurobiology, Keck School Of Medicine, University Of Southern California, Los Angeles, California
| | - Li I Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University Of Southern California, Los Angeles, California, 90033.,Department of Physiology and Biophysics, Keck School Of Medicine, University Of Southern California, Los Angeles, California
| |
Collapse
|
48
|
Fritzsch B, Pan N, Jahan I, Elliott KL. Inner ear development: building a spiral ganglion and an organ of Corti out of unspecified ectoderm. Cell Tissue Res 2015; 361:7-24. [PMID: 25381571 PMCID: PMC4426086 DOI: 10.1007/s00441-014-2031-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 10/09/2014] [Indexed: 01/21/2023]
Abstract
The mammalian inner ear develops from a placodal thickening into a complex labyrinth of ducts with five sensory organs specialized to detect position and movement in space. The mammalian ear also develops a spiraled cochlear duct containing the auditory organ, the organ of Corti (OC), specialized to translate sound into hearing. Development of the OC from a uniform sheet of ectoderm requires unparalleled precision in the topological developmental engineering of four different general cell types, namely sensory neurons, hair cells, supporting cells, and general otic epithelium, into a mosaic of ten distinctly recognizable cell types in and around the OC, each with a unique distribution. Moreover, the OC receives unique innervation by ear-derived spiral ganglion afferents and brainstem-derived motor neurons as efferents and requires neural-crest-derived Schwann cells to form myelin and neural-crest-derived cells to induce the stria vascularis. This transformation of a sheet of cells into a complicated interdigitating set of cells necessitates the orchestrated expression of multiple transcription factors that enable the cellular transformation from ectoderm into neurosensory cells forming the spiral ganglion neurons (SGNs), while simultaneously transforming the flat epithelium into a tube, the cochlear duct, housing the OC. In addition to the cellular and conformational changes forming the cochlear duct with the OC, changes in the surrounding periotic mesenchyme form passageways for sound to stimulate the OC. We review molecular developmental data, generated predominantly in mice, in order to integrate the well-described expression changes of transcription factors and their actions, as revealed in mutants, in the formation of SGNs and OC in the correct position and orientation with suitable innervation. Understanding the molecular basis of these developmental changes leading to the formation of the mammalian OC and highlighting the gaps in our knowledge might guide in vivo attempts to regenerate this most complicated cellular mosaic of the mammalian body for the reconstitution of hearing in a rapidly growing population of aging people suffering from hearing loss.
Collapse
Affiliation(s)
- Bernd Fritzsch
- College of Liberal Arts and Sciences, Department of Biology, University of Iowa, 143 BB, 123 Jefferson Avenue, Iowa City, IA 52242, USA,
| | | | | | | |
Collapse
|
49
|
Wan G, Corfas G. No longer falling on deaf ears: mechanisms of degeneration and regeneration of cochlear ribbon synapses. Hear Res 2015; 329:1-10. [PMID: 25937135 DOI: 10.1016/j.heares.2015.04.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 04/01/2015] [Accepted: 04/20/2015] [Indexed: 01/02/2023]
Abstract
Cochlear ribbon synapses are required for the rapid and precise neural transmission of acoustic signals from inner hair cells to the spiral ganglion neurons. Emerging evidence suggests that damage to these synapses represents an important form of cochlear neuropathy that might be highly prevalent in sensorineural hearing loss. In this review, we discuss our current knowledge on how ribbon synapses are damaged by noise and during aging, as well as potential strategies to promote ribbon synapse regeneration for hearing restoration.
Collapse
Affiliation(s)
- Guoqiang Wan
- Kresge Hearing Research Institute, Department of Otolaryngology - Head and Neck Surgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Gabriel Corfas
- Kresge Hearing Research Institute, Department of Otolaryngology - Head and Neck Surgery, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
50
|
Defourny J, Mateo Sánchez S, Schoonaert L, Robberecht W, Davy A, Nguyen L, Malgrange B. Cochlear supporting cell transdifferentiation and integration into hair cell layers by inhibition of ephrin-B2 signalling. Nat Commun 2015; 6:7017. [DOI: 10.1038/ncomms8017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 03/25/2015] [Indexed: 01/08/2023] Open
|