1
|
Jaime Tobón LM, Moser T. Bridging the gap between presynaptic hair cell function and neural sound encoding. eLife 2024; 12:RP93749. [PMID: 39718472 DOI: 10.7554/elife.93749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024] Open
Abstract
Neural diversity can expand the encoding capacity of a circuitry. A striking example of diverse structure and function is presented by the afferent synapses between inner hair cells (IHCs) and spiral ganglion neurons (SGNs) in the cochlea. Presynaptic active zones at the pillar IHC side activate at lower IHC potentials than those of the modiolar side that have more presynaptic Ca2+ channels. The postsynaptic SGNs differ in their spontaneous firing rates, sound thresholds, and operating ranges. While a causal relationship between synaptic heterogeneity and neural response diversity seems likely, experimental evidence linking synaptic and SGN physiology has remained difficult to obtain. Here, we aimed at bridging this gap by ex vivo paired recordings of murine IHCs and postsynaptic SGN boutons with stimuli and conditions aimed to mimic those of in vivo SGN characterization. Synapses with high spontaneous rate of release (SR) were found predominantly on the pillar side of the IHC. These high SR synapses had larger and more temporally compact spontaneous EPSCs, lower voltage thresholds, tighter coupling of Ca2+ channels and vesicular release sites, shorter response latencies, and higher initial release rates. This study indicates that synaptic heterogeneity in IHCs directly contributes to the diversity of spontaneous and sound-evoked firing of SGNs.
Collapse
Affiliation(s)
- Lina María Jaime Tobón
- Auditory Neuroscience and Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany
- Collaborative Research Center, University of Göttingen, Göttingen, Germany
- Cluster of Excellence 'Multiscale Bioimaging of Excitable Cells', Göttingen, Germany
| | - Tobias Moser
- Auditory Neuroscience and Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany
- Collaborative Research Center, University of Göttingen, Göttingen, Germany
- Cluster of Excellence 'Multiscale Bioimaging of Excitable Cells', Göttingen, Germany
| |
Collapse
|
2
|
Kostrikov S, Hjortkjaer J, Dau T, Corfas G, Liberman LD, Liberman MC. A modiolar-pillar gradient in auditory-nerve dendritic length: A novel post-synaptic contribution to dynamic range? Hear Res 2024; 456:109172. [PMID: 39708764 DOI: 10.1016/j.heares.2024.109172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/06/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Auditory-nerve fibers (ANFs) from a given cochlear region can vary in threshold sensitivity by up to 60 dB, corresponding to a 1000-fold difference in stimulus level, although each fiber innervates a single inner hair cell (IHC) via a single synapse. ANFs with high-thresholds also have low spontaneous rates (SRs) and synapse on the side of the IHC closer to the modiolus, whereas the low-threshold, high-SR fibers synapse on the side closer to the pillar cells. Prior biophysical work has identified modiolar-pillar differences in both pre- and post-synaptic properties, but a comprehensive explanation for the wide range of sensitivities remains elusive. Here, in guinea pigs, we used immunostaining for several neuronal markers, including Caspr, a key protein in nodes of Ranvier, to reveal a novel modiolar-pillar gradient in the location of the first ANF heminodes, presumed to be the site of the spike generator, just outside the sensory epithelium. Along the cochlea, from apex to base, the unmyelinated terminal dendrites of modiolar ANFs were 2-4 times longer than those of pillar ANFs. This modiolar-pillar gradient in dendritic length, coupled with the 2-4 fold smaller caliber of modiolar dendrites seen in prior single-fiber labeling studies, suggests there could be a large difference in the number of length constants between the synapse and the spike initiation zone for low- vs high-SR fibers. The resultant differences in attenuation of post-synaptic potentials propagating along these unmyelinated dendrites could be a key contributor to the observed range of threshold sensitivities among ANFs.
Collapse
Affiliation(s)
- Serhii Kostrikov
- Department of Health Technology, Centre for Auditory Neuroscience, Hearing Systems, Technical University of Denmark, Lyngby, Denmark
| | - Jens Hjortkjaer
- Department of Health Technology, Centre for Auditory Neuroscience, Hearing Systems, Technical University of Denmark, Lyngby, Denmark
| | - Torsten Dau
- Department of Health Technology, Centre for Auditory Neuroscience, Hearing Systems, Technical University of Denmark, Lyngby, Denmark
| | - Gabriel Corfas
- Department of Otolaryngology-Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI, United States
| | - Leslie D Liberman
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA, United States
| | - M Charles Liberman
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA, United States; Department of Otolaryngology-Head & Neck Surgery, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
3
|
Kreeger LJ, Honnuraiah S, Maeker S, Shea S, Fishell G, Goodrich LV. An Anatomical and Physiological Basis for Flexible Coincidence Detection in the Auditory System. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.29.582808. [PMID: 38464181 PMCID: PMC10925315 DOI: 10.1101/2024.02.29.582808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Animals navigate the auditory world by recognizing complex sounds, from the rustle of a predator to the call of a potential mate. This ability depends in part on the octopus cells of the auditory brainstem, which respond to multiple frequencies that change over time, as occurs in natural stimuli. Unlike the average neuron, which integrates inputs over time on the order of tens of milliseconds, octopus cells must detect momentary coincidence of excitatory inputs from the cochlea during an ongoing sound on both the millisecond and submillisecond time scale. Here, we show that octopus cells receive inhibitory inputs on their dendrites that enhance opportunities for coincidence detection in the cell body, thereby allowing for responses both to rapid onsets at the beginning of a sound and to frequency modulations during the sound. This mechanism is crucial for the fundamental process of integrating the synchronized frequencies of natural auditory signals over time.
Collapse
Affiliation(s)
- Lauren J Kreeger
- Harvard Medical School, Department of Neurobiology, Boston, MA 02115, USA
| | - Suraj Honnuraiah
- Harvard Medical School, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sydney Maeker
- Harvard Medical School, Department of Neurobiology, Boston, MA 02115, USA
| | - Siobhan Shea
- Harvard Medical School, Department of Neurobiology, Boston, MA 02115, USA
| | - Gord Fishell
- Harvard Medical School, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Lisa V Goodrich
- Harvard Medical School, Department of Neurobiology, Boston, MA 02115, USA
| |
Collapse
|
4
|
Kostrikov S, Hjortkjaer J, Dau T, Corfas G, Liberman LD, Liberman MC. A modiolar-pillar gradient in auditory-nerve dendritic length: a novel post-synaptic contribution to dynamic range? BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.04.621861. [PMID: 39574647 PMCID: PMC11580876 DOI: 10.1101/2024.11.04.621861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2024]
Abstract
Auditory-nerve fibers (ANFs) from a given cochlear region can vary in threshold sensitivity by up to 60 dB, corresponding to a 1000-fold difference in stimulus level, although each fiber innervates a single inner hair cell (IHC) via a single synapse. ANFs with high-thresholds also have low spontaneous rates (SRs) and synapse on the side of the IHC closer to the modiolus, whereas the low-threshold, high-SR fibers synapse on the side closer to the pillar cells. Prior biophysical work has identified modiolar-pillar differences in both pre- and post-synaptic properties, but a comprehensive explanation for the wide range of sensitivities remains elusive. Here, in guinea pigs, we used immunostaining for several neuronal markers, including Caspr, a key protein in nodes of Ranvier, to reveal a novel modiolar-pillar gradient in the location of the first ANF heminodes, presumed to be the site of the spike generator, just outside the sensory epithelium. Along the cochlea, from apex to base, the unmyelinated terminal dendrites of modiolar ANFs were 2 - 4 times longer than those of pillar ANFs. This modiolar-pillar gradient in dendritic length, coupled with the 2 - 4 fold smaller caliber of modiolar dendrites seen in prior single-fiber labeling studies, suggests there could be a large difference in the number of length constants between the synapse and the spike initiation zone for low- vs high-SR fibers. The resultant differences in attenuation of post-synaptic potentials propagating along these unmyelinated dendrites could be a key contributor to the observed range of threshold sensitivities among ANFs.
Collapse
|
5
|
Hosoya M, Ueno M, Shimanuki MN, Nishiyama T, Oishi N, Ozawa H. A primate model animal revealed the inter-species differences and similarities in the subtype specifications of the spiral ganglion neurons. Sci Rep 2024; 14:25166. [PMID: 39448766 PMCID: PMC11502759 DOI: 10.1038/s41598-024-76892-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024] Open
Abstract
Type I spiral ganglion neurons are peripheral neurons essential for hearing perception. While they can be subdivided in mice based on characteristic gene expression patterns, detailed examinations of these subtypes in primates and humans are lacking. In this study, we investigated the developmental subtypes of spiral ganglion neurons in the common marmoset (Callithrix jacchus). We confirmed that Type I spiral ganglion can be divided based on the characteristic gene expression patterns of several marker genes. However, some combinations of these genes differ from those in rodents, suggesting common marmoset's suitability for advancing our understanding of human cochlear development. Additionally, identifying the essential time points for subtype specifications and subsequent maturation will aid in studying the primate-specific developmental biology of the inner ear. This could lead to new treatment strategies for hearing loss in humans and be valuable for studying age-related hearing loss, as well as designing regenerative therapies.
Collapse
Affiliation(s)
- Makoto Hosoya
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Masafumi Ueno
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Marie N Shimanuki
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Takanori Nishiyama
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Naoki Oishi
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hiroyuki Ozawa
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| |
Collapse
|
6
|
Tao J, Gu Y, Zhang Z, Weng G, Liu Y, Ren J, Shi Y, Qiu J, Wang Y, Su D, Wang R, Fu Y, Liu T, Ye L, Luo W, Chen H, Yang G, Cao Z, Huang H, Xiao J, Ren B, You L, Zhang T, Zhao Y. CALB2 drives pancreatic cancer metastasis through inflammatory reprogramming of the tumor microenvironment. J Exp Clin Cancer Res 2024; 43:277. [PMID: 39358777 PMCID: PMC11448066 DOI: 10.1186/s13046-024-03201-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Early dissemination to distant organs accounts for the dismal prognosis of patients with pancreatic ductal adenocarcinoma (PDAC). Chronic, dysregulated, persistent and unresolved inflammation provides a preferred tumor microenvironment (TME) for tumorigenesis, development, and metastasis. A better understanding of the key regulators that maintain inflammatory TME and the development of predictive biomarkers to identify patients who are most likely to benefit from specific inflammatory-targeted therapies is crucial for advancing personalized cancer treatment. METHODS This study identified cell-specific expression of CALB2 in human PDAC through single-cell RNA sequencing analysis and assessed its clinicopathological correlations in tissue microarray using multi-color immunofluorescence. Co-culture systems containing cancer-associated fibroblasts (CAFs) and patient-derived organoids (PDOs) in vitro and in vivo were employed to elucidate the effects of CALB2-activated CAFs on PDAC malignancy. Furthermore, CUT&RUN assays, luciferase reporter assays, RNA sequencing, and gain- or loss-of-function assays were used to unravel the molecular mechanisms of CALB2-mediated inflammatory reprogramming and metastasis. Additionally, immunocompetent KPC organoid allograft models were constructed to evaluate CALB2-induced immunosuppression and PDAC metastasis, as well as the efficacy of inflammation-targeted therapy. RESULTS CALB2 was highly expressed both in CAFs and cancer cells and correlated with an unfavorable prognosis and immunosuppressive TME in PDAC patients. CALB2 collaborated with hypoxia to activate an inflammatory fibroblast phenotype, which promoted PDAC cell migration and PDO growth in vitro and in vivo. In turn, CALB2-activated CAFs upregulated CALB2 expression in cancer cells through IL6-STAT3 signaling-mediated direct transcription. In cancer cells, CALB2 further activated Ca2+-CXCL14 inflammatory axis to facilitate PDAC metastatic outgrowth and immunosuppression. Genetic or pharmaceutical inhibition of CXCL14 significantly suppressed CALB2-mediated metastatic colonization of PDAC cells in vivo and extended mouse survival. CONCLUSIONS These findings identify CALB2 as a key regulator of inflammatory reprogramming to promote PDAC metastatic progression. Combination therapy with αCXCL14 monoclonal antibody and gemcitabine emerges as a promising strategy to suppress distant metastasis and improve survival outcomes in PDAC with CALB2 overexpression.
Collapse
Affiliation(s)
- Jinxin Tao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yani Gu
- Institute of Clinical Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Translational Medicine Center, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Zeyu Zhang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Guihu Weng
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yueze Liu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jie Ren
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yanan Shi
- Biomedical Engineering Facility of National Infrastructures for Translational Medicine, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jiangdong Qiu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Yuanyang Wang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Dan Su
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ruobing Wang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
- Department of General Surgery, China‑Japan Friendship Hospital, Beijing, 100029, China
| | - Yifan Fu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Tao Liu
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Liyuan Ye
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Wenhao Luo
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Hao Chen
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Gang Yang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zhe Cao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Hua Huang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Jianchun Xiao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Bo Ren
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Lei You
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Taiping Zhang
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Yupei Zhao
- General Surgery Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
7
|
Conrad LJ, Grandi FC, Carlton AJ, Jeng JY, de Tomasi L, Zarecki P, Marcotti W, Johnson SL, Mustapha M. The upregulation of K + and HCN channels in developing spiral ganglion neurons is mediated by cochlear inner hair cells. J Physiol 2024; 602:5329-5351. [PMID: 39324853 DOI: 10.1113/jp286134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 09/02/2024] [Indexed: 09/27/2024] Open
Abstract
Spiral ganglion neurons (SGNs) are primary sensory afferent neurons that relay acoustic information from the cochlear inner hair cells (IHCs) to the brainstem. The response properties of different SGNs diverge to represent a wide range of sound intensities in an action-potential code. This biophysical heterogeneity is established during pre-hearing stages of development, a time when IHCs fire spontaneous Ca2+ action potentials that drive glutamate release from their ribbon synapses onto the SGN terminals. The role of spontaneous IHC activity in the refinement of SGN characteristics is still largely unknown. Using pre-hearing otoferlin knockout mice (Otof-/-), in which Ca2+-dependent exocytosis in IHCs is abolished, we found that developing SGNs fail to upregulate low-voltage-activated K+-channels and hyperpolarisation-activated cyclic-nucleotide-gated channels. This delayed maturation resulted in hyperexcitable SGNs with immature firing characteristics. We have also shown that SGNs that synapse with the pillar side of the IHCs selectively express a resurgent K+ current, highlighting a novel biophysical marker for these neurons. RNA-sequencing showed that several K+ channels are downregulated in Otof-/- mice, further supporting the electrophysiological recordings. Our data demonstrate that spontaneous Ca2+-dependent activity in pre-hearing IHCs regulates some of the key biophysical and molecular features of the developing SGNs. KEY POINTS: Ca2+-dependent exocytosis in inner hair cells (IHCs) is otoferlin-dependent as early as postnatal day 1. A lack of otoferlin in IHCs affects potassium channel expression in SGNs. The absence of otoferlin is associated with SGN hyperexcitability. We propose that type I spiral ganglion neuron functional maturation depends on IHC exocytosis.
Collapse
Affiliation(s)
- Linus J Conrad
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - Fiorella C Grandi
- INSERM, Institute de Myologie, Centre de Recherche en Myologie F-75013, Sorbonne Université, Paris, France
| | - Adam J Carlton
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - Jing-Yi Jeng
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - Lara de Tomasi
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - Patryk Zarecki
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - Walter Marcotti
- School of Biosciences, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Stuart L Johnson
- School of Biosciences, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Mirna Mustapha
- School of Biosciences, University of Sheffield, Sheffield, UK
- Neuroscience Institute, University of Sheffield, Sheffield, UK
| |
Collapse
|
8
|
Pyott SJ, Pavlinkova G, Yamoah EN, Fritzsch B. Harmony in the Molecular Orchestra of Hearing: Developmental Mechanisms from the Ear to the Brain. Annu Rev Neurosci 2024; 47:1-20. [PMID: 38360566 DOI: 10.1146/annurev-neuro-081423-093942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Auditory processing in mammals begins in the peripheral inner ear and extends to the auditory cortex. Sound is transduced from mechanical stimuli into electrochemical signals of hair cells, which relay auditory information via the primary auditory neurons to cochlear nuclei. Information is subsequently processed in the superior olivary complex, lateral lemniscus, and inferior colliculus and projects to the auditory cortex via the medial geniculate body in the thalamus. Recent advances have provided valuable insights into the development and functioning of auditory structures, complementing our understanding of the physiological mechanisms underlying auditory processing. This comprehensive review explores the genetic mechanisms required for auditory system development from the peripheral cochlea to the auditory cortex. We highlight transcription factors and other genes with key recurring and interacting roles in guiding auditory system development and organization. Understanding these gene regulatory networks holds promise for developing novel therapeutic strategies for hearing disorders, benefiting millions globally.
Collapse
Affiliation(s)
- Sonja J Pyott
- Department of Otorhinolaryngology and Head and Neck Surgery, University Medical Center Groningen, Graduate School of Medical Sciences, and Research School of Behavioral and Cognitive Neurosciences, University of Groningen, Groningen, The Netherlands
| | - Gabriela Pavlinkova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology, Czech Academy of Sciences, Vestec, Czechia
| | - Ebenezer N Yamoah
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, Nevada, USA
| | - Bernd Fritzsch
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA;
| |
Collapse
|
9
|
Kersbergen CJ, Bergles DE. Priming central sound processing circuits through induction of spontaneous activity in the cochlea before hearing onset. Trends Neurosci 2024; 47:522-537. [PMID: 38782701 PMCID: PMC11236524 DOI: 10.1016/j.tins.2024.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 04/02/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024]
Abstract
Sensory systems experience a period of intrinsically generated neural activity before maturation is complete and sensory transduction occurs. Here we review evidence describing the mechanisms and functions of this 'spontaneous' activity in the auditory system. Both ex vivo and in vivo studies indicate that this correlated activity is initiated by non-sensory supporting cells within the developing cochlea, which induce depolarization and burst firing of groups of nearby hair cells in the sensory epithelium, activity that is conveyed to auditory neurons that will later process similar sound features. This stereotyped neural burst firing promotes cellular maturation, synaptic refinement, acoustic sensitivity, and establishment of sound-responsive domains in the brain. While sensitive to perturbation, the developing auditory system exhibits remarkable homeostatic mechanisms to preserve periodic burst firing in deaf mice. Preservation of this early spontaneous activity in the context of deafness may enhance the efficacy of later interventions to restore hearing.
Collapse
Affiliation(s)
- Calvin J Kersbergen
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - Dwight E Bergles
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA; Department of Otolaryngology - Head and Neck Surgery, Johns Hopkins University, Baltimore, MD, USA; Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
10
|
Carlton AJ, Jeng JY, Grandi FC, De Faveri F, Amariutei AE, De Tomasi L, O'Connor A, Johnson SL, Furness DN, Brown SDM, Ceriani F, Bowl MR, Mustapha M, Marcotti W. BAI1 localizes AMPA receptors at the cochlear afferent post-synaptic density and is essential for hearing. Cell Rep 2024; 43:114025. [PMID: 38564333 DOI: 10.1016/j.celrep.2024.114025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 01/25/2024] [Accepted: 03/15/2024] [Indexed: 04/04/2024] Open
Abstract
Type I spiral ganglion neurons (SGNs) convey sound information to the central auditory pathway by forming synapses with inner hair cells (IHCs) in the mammalian cochlea. The molecular mechanisms regulating the formation of the post-synaptic density (PSD) in the SGN afferent terminals are still unclear. Here, we demonstrate that brain-specific angiogenesis inhibitor 1 (BAI1) is required for the clustering of AMPA receptors GluR2-4 (glutamate receptors 2-4) at the PSD. Adult Bai1-deficient mice have functional IHCs but fail to transmit information to the SGNs, leading to highly raised hearing thresholds. Despite the almost complete absence of AMPA receptor subunits, the SGN fibers innervating the IHCs do not degenerate. Furthermore, we show that AMPA receptors are still expressed in the cochlea of Bai1-deficient mice, highlighting a role for BAI1 in trafficking or anchoring GluR2-4 to the PSDs. These findings identify molecular and functional mechanisms required for sound encoding at cochlear ribbon synapses.
Collapse
Affiliation(s)
- Adam J Carlton
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| | - Jing-Yi Jeng
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| | - Fiorella C Grandi
- Sorbonne Université, INSERM, Institute de Myologie, Centre de Recherche en Myologie, 75013 Paris, France
| | | | - Ana E Amariutei
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| | - Lara De Tomasi
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| | - Andrew O'Connor
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| | - Stuart L Johnson
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK; Neuroscience Institute, University of Sheffield, Sheffield S10 2TN, UK
| | - David N Furness
- School of Life Sciences, Keele University, Keele ST5 5BG, UK
| | - Steve D M Brown
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK
| | - Federico Ceriani
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
| | - Michael R Bowl
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK
| | - Mirna Mustapha
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK; Neuroscience Institute, University of Sheffield, Sheffield S10 2TN, UK
| | - Walter Marcotti
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK; Neuroscience Institute, University of Sheffield, Sheffield S10 2TN, UK.
| |
Collapse
|
11
|
Xie R, Wang M, Zhang C. Mechanisms of age-related hearing loss at the auditory nerve central synapses and postsynaptic neurons in the cochlear nucleus. Hear Res 2024; 442:108935. [PMID: 38113793 PMCID: PMC10842789 DOI: 10.1016/j.heares.2023.108935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 12/21/2023]
Abstract
Sound information is transduced from mechanical vibration to electrical signals in the cochlea, conveyed to and further processed in the brain to form auditory perception. During the process, spiral ganglion neurons (SGNs) are the key cells that connect the peripheral and central auditory systems by receiving information from hair cells in the cochlea and transmitting it to neurons of the cochlear nucleus (CN). Decades of research in the cochlea greatly improved our understanding of SGN function under normal and pathological conditions, especially about the roles of different subtypes of SGNs and their peripheral synapses. However, it remains less clear how SGN central terminals or auditory nerve (AN) synapses connect to CN neurons, and ultimately how peripheral pathology links to structural alterations and functional deficits in the central auditory nervous system. This review discusses recent progress about the morphological and physiological properties of different subtypes of AN synapses and associated postsynaptic CN neurons, their changes during aging, and the potential mechanisms underlying age-related hearing loss.
Collapse
Affiliation(s)
- Ruili Xie
- Department of Otolaryngology, The Ohio State University, 420 W 12th Ave, Columbus OH 43210, USA; Department of Neuroscience, The Ohio State University, 420W 12th Ave, Columbus, OH 43210, USA.
| | - Meijian Wang
- Department of Otolaryngology, The Ohio State University, 420 W 12th Ave, Columbus OH 43210, USA
| | - Chuangeng Zhang
- Department of Otolaryngology, The Ohio State University, 420 W 12th Ave, Columbus OH 43210, USA
| |
Collapse
|
12
|
Jaime Tobón LM, Moser T. Ca 2+ regulation of glutamate release from inner hair cells of hearing mice. Proc Natl Acad Sci U S A 2023; 120:e2311539120. [PMID: 38019860 DOI: 10.1073/pnas.2311539120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
In our hearing organ, sound is encoded at ribbon synapses formed by inner hair cells (IHCs) and spiral ganglion neurons (SGNs). How the underlying synaptic vesicle (SV) release is controlled by Ca2+ in IHCs of hearing animals remained to be investigated. Here, we performed patch-clamp SGN recordings of the initial rate of release evoked by brief IHC Ca2+-influx in an ex vivo cochlear preparation from hearing mice. We aimed to closely mimic physiological conditions by perforated-patch recordings from IHCs kept at the physiological resting potential and at body temperature. We found release to relate supralinearly to Ca2+-influx (power, m: 4.3) when manipulating the [Ca2+] available for SV release by Zn2+-flicker-blocking of the single Ca2+-channel current. In contrast, a near linear Ca2+ dependence (m: 1.2 to 1.5) was observed when varying the number of open Ca2+-channels during deactivating Ca2+-currents and by dihydropyridine channel-inhibition. Concurrent changes of number and current of open Ca2+-channels over the range of physiological depolarizations revealed m: 1.8. These findings indicate that SV release requires ~4 Ca2+-ions to bind to their Ca2+-sensor of fusion. We interpret the near linear Ca2+-dependence of release during manipulations that change the number of open Ca2+-channels to reflect control of SV release by the high [Ca2+] in the Ca2+-nanodomain of one or few nearby Ca2+-channels. We propose that a combination of Ca2+ nanodomain control and supralinear intrinsic Ca2+-dependence of fusion optimally links SV release to the timing and amplitude of the IHC receptor potential and separates it from other IHC Ca2+-signals unrelated to afferent synaptic transmission.
Collapse
Affiliation(s)
- Lina María Jaime Tobón
- Auditory Neuroscience and Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
- Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen 37075, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen 37075, Germany
- Multiscale Bioimaging of Excitable Cells, Cluster of Excellence, Göttingen 37075, Germany
| | - Tobias Moser
- Auditory Neuroscience and Synaptic Nanophysiology Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
- Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen 37075, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen 37075, Germany
- Multiscale Bioimaging of Excitable Cells, Cluster of Excellence, Göttingen 37075, Germany
| |
Collapse
|
13
|
Moser T, Karagulyan N, Neef J, Jaime Tobón LM. Diversity matters - extending sound intensity coding by inner hair cells via heterogeneous synapses. EMBO J 2023; 42:e114587. [PMID: 37800695 PMCID: PMC10690447 DOI: 10.15252/embj.2023114587] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/26/2023] [Accepted: 08/07/2023] [Indexed: 10/07/2023] Open
Abstract
Our sense of hearing enables the processing of stimuli that differ in sound pressure by more than six orders of magnitude. How to process a wide range of stimulus intensities with temporal precision is an enigmatic phenomenon of the auditory system. Downstream of dynamic range compression by active cochlear micromechanics, the inner hair cells (IHCs) cover the full intensity range of sound input. Yet, the firing rate in each of their postsynaptic spiral ganglion neurons (SGNs) encodes only a fraction of it. As a population, spiral ganglion neurons with their respective individual coding fractions cover the entire audible range. How such "dynamic range fractionation" arises is a topic of current research and the focus of this review. Here, we discuss mechanisms for generating the diverse functional properties of SGNs and formulate testable hypotheses. We postulate that an interplay of synaptic heterogeneity, molecularly distinct subtypes of SGNs, and efferent modulation serves the neural decomposition of sound information and thus contributes to a population code for sound intensity.
Collapse
Affiliation(s)
- Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLabUniversity Medical Center GöttingenGöttingenGermany
- Auditory Neuroscience and Synaptic Nanophysiology GroupMax Planck Institute for Multidisciplinary SciencesGöttingenGermany
- Cluster of Excellence “Multiscale Bioimaging of Excitable Cells”GöttingenGermany
| | - Nare Karagulyan
- Institute for Auditory Neuroscience and InnerEarLabUniversity Medical Center GöttingenGöttingenGermany
- Auditory Neuroscience and Synaptic Nanophysiology GroupMax Planck Institute for Multidisciplinary SciencesGöttingenGermany
- Hertha Sponer CollegeCluster of Excellence “Multiscale Bioimaging of Excitable Cells” Cluster of ExcellenceGöttingenGermany
| | - Jakob Neef
- Institute for Auditory Neuroscience and InnerEarLabUniversity Medical Center GöttingenGöttingenGermany
- Auditory Neuroscience and Synaptic Nanophysiology GroupMax Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Lina María Jaime Tobón
- Institute for Auditory Neuroscience and InnerEarLabUniversity Medical Center GöttingenGöttingenGermany
- Auditory Neuroscience and Synaptic Nanophysiology GroupMax Planck Institute for Multidisciplinary SciencesGöttingenGermany
- Hertha Sponer CollegeCluster of Excellence “Multiscale Bioimaging of Excitable Cells” Cluster of ExcellenceGöttingenGermany
| |
Collapse
|
14
|
Boussaty EC, Tedeschi N, Novotny M, Ninoyu Y, Du E, Draf C, Zhang Y, Manor U, Scheuermann RH, Friedman R. Cochlear transcriptome analysis of an outbred mouse population (CFW). Front Cell Neurosci 2023; 17:1256619. [PMID: 38094513 PMCID: PMC10716316 DOI: 10.3389/fncel.2023.1256619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/11/2023] [Indexed: 12/20/2023] Open
Abstract
Age-related hearing loss (ARHL) is the most common cause of hearing loss and one of the most prevalent conditions affecting the elderly worldwide. Despite evidence from our lab and others about its polygenic nature, little is known about the specific genes, cell types, and pathways involved in ARHL, impeding the development of therapeutic interventions. In this manuscript, we describe, for the first time, the complete cell-type specific transcriptome of the aging mouse cochlea using snRNA-seq in an outbred mouse model in relation to auditory threshold variation. Cochlear cell types were identified using unsupervised clustering and annotated via a three-tiered approach-first by linking to expression of known marker genes, then using the NSForest algorithm to select minimum cluster-specific marker genes and reduce dimensional feature space for statistical comparison of our clusters with existing publicly-available data sets on the gEAR website, and finally, by validating and refining the annotations using Multiplexed Error Robust Fluorescence In Situ Hybridization (MERFISH) and the cluster-specific marker genes as probes. We report on 60 unique cell-types expanding the number of defined cochlear cell types by more than two times. Importantly, we show significant specific cell type increases and decreases associated with loss of hearing acuity implicating specific subsets of hair cell subtypes, ganglion cell subtypes, and cell subtypes within the stria vascularis in this model of ARHL. These results provide a view into the cellular and molecular mechanisms responsible for age-related hearing loss and pathways for therapeutic targeting.
Collapse
Affiliation(s)
- Ely Cheikh Boussaty
- Department of Otolaryngology, University of California, San Diego, La Jolla, CA, United States
| | - Neil Tedeschi
- J. Craig Venter Institute, La Jolla, CA, United States
| | - Mark Novotny
- J. Craig Venter Institute, La Jolla, CA, United States
| | - Yuzuru Ninoyu
- Department of Otolaryngology, University of California, San Diego, La Jolla, CA, United States
| | - Eric Du
- Department of Otolaryngology, University of California, San Diego, La Jolla, CA, United States
| | - Clara Draf
- Department of Otolaryngology, University of California, San Diego, La Jolla, CA, United States
| | - Yun Zhang
- J. Craig Venter Institute, La Jolla, CA, United States
| | - Uri Manor
- Department of Cell and Developmental Biology, University of California San Diego, Salk Institute for Biological Studies, Waitt Advanced Biophotonics Center, La Jolla, CA, United States
| | - Richard H. Scheuermann
- J. Craig Venter Institute, La Jolla, CA, United States
- Department of Pathology, University of California, San Diego, La Jolla, CA, United States
| | - Rick Friedman
- Department of Otolaryngology, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
15
|
Karagulyan N, Moser T. Synaptic activity is not required for establishing heterogeneity of inner hair cell ribbon synapses. Front Mol Neurosci 2023; 16:1248941. [PMID: 37745283 PMCID: PMC10512025 DOI: 10.3389/fnmol.2023.1248941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/17/2023] [Indexed: 09/26/2023] Open
Abstract
Neural sound encoding in the mammalian cochlea faces the challenge of representing audible sound pressures that vary over six orders of magnitude. The cochlea meets this demand through the use of active micromechanics as well as the diversity and adaptation of afferent neurons and their synapses. Mechanisms underlying neural diversity likely include heterogeneous presynaptic input from inner hair cells (IHCs) to spiral ganglion neurons (SGNs) as well as differences in the molecular profile of SGNs and in their efferent control. Here, we tested whether glutamate release from IHCs, previously found to be critical for maintaining different molecular SGN profiles, is required for establishing heterogeneity of active zones (AZs) in IHCs. We analyzed structural and functional heterogeneity of IHC AZs in mouse mutants with disrupted glutamate release from IHCs due to lack of a vesicular glutamate transporter (Vglut3) or impaired exocytosis due to defective otoferlin. We found the variance of the voltage-dependence of presynaptic Ca2+ influx to be reduced in exocytosis-deficient IHCs of otoferlin mutants. Yet, the spatial gradients of maximal amplitude and voltage-dependence of Ca2+ influx along the pillar-modiolar IHC axis were maintained in both mutants. Further immunohistochemical analysis showed an intact spatial gradient of ribbon size in Vglut3-/- mice. These results indicate that IHC exocytosis and glutamate release are not strictly required for establishing the heterogeneity of IHC AZs.
Collapse
Affiliation(s)
- Nare Karagulyan
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience and Nanophysiology Group, Max Planck Institute of Multidisciplinary Sciences, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
- Göttingen Graduate School for Neurosciences and Molecular Biosciences, University of Göttingen, Göttingen, Germany
- Hertha Sponer College, Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, Göttingen, Germany
| | - Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience and Nanophysiology Group, Max Planck Institute of Multidisciplinary Sciences, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
- Göttingen Graduate School for Neurosciences and Molecular Biosciences, University of Göttingen, Göttingen, Germany
- Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|