1
|
Yang YM, Fekete A, Arsenault J, Sengar AS, Aitoubah J, Grande G, Li A, Salter EW, Wang A, Mark MD, Herlitze S, Egan SE, Salter MW, Wang LY. Intersectin-1 enhances calcium-dependent replenishment of the readily releasable pool of synaptic vesicles during development. J Physiol 2024. [PMID: 39383250 DOI: 10.1113/jp286462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/06/2024] [Indexed: 10/11/2024] Open
Abstract
Intersectin-1 (Itsn1) is a scaffold protein that plays a key role in coupling exocytosis and endocytosis of synaptic vesicles (SVs). However, it is unclear whether and how Itsn1 regulates these processes to support efficient neurotransmission during development. To address this, we examined the calyx of Held synapse in the auditory brainstem of wild-type and Itsn1 mutant mice before (immature) and after (mature) the onset of hearing. Itsn1 was present in the pre- and postsynaptic compartments at both developmental stages. Loss of function of Itsn1 did not alter presynaptic action potentials, Ca2+ entry via voltage-gated Ca2+ channels (VGCCs), transmitter release or short-term depression (STD) induced by depletion of SVs in the readily releasable pool (RRP) in either age group. Yet, fast Ca2+-dependent recovery from STD was attenuated in mature mutant synapses, while it was unchanged in immature mutant synapses. This deficit at mature synapses was rescued by introducing the DH-PH domains of Itsn1 into the presynaptic terminals. Inhibition of dynamin, which interacts with Itsn1 during endocytosis, had no effect on STD recovery. Interestingly, we found a developmental enrichment of Itsn1 near VGCCs, which may underlie the Itsn1-mediated fast replenishment of the RRP. Consequently, the absence of Itsn1 in mature synapses led to a higher failure rate of postsynaptic spiking during high-frequency synaptic transmission. Taken together, our findings suggest that Itsn1 translocation to the vicinity of VGCCs during development is crucial for accelerating Ca2+-dependent RRP replenishment and sustaining high-fidelity neurotransmission. KEY POINTS: Itsn1 is expressed in the pre- and postsynaptic compartments of the calyx of Held synapse. Developmental upregulation of vesicular glutamate transporter-1 is Itsn1 dependent. Itsn1 does not affect basal synaptic transmission at different developmental stages. Itsn1 is required for Ca2+-dependent recovery from short-term depression in mature synapses. Itsn1 mediates the recovery through its DH-PH domains, independent of its interactive partner dynamin. Itsn1 translocates to the vicinity of presynaptic Ca2+ channels during development. Itsn1 supports high-fidelity neurotransmission by enabling rapid recovery from vesicular depletion during repetitive activity.
Collapse
Affiliation(s)
- Yi-Mei Yang
- Neurosciences and Mental Health, SickKids Research Institute, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Biomedical Sciences, University of Minnesota, Duluth, Minnesota, USA
| | - Adam Fekete
- Neurosciences and Mental Health, SickKids Research Institute, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Jason Arsenault
- Neurosciences and Mental Health, SickKids Research Institute, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Ameet S Sengar
- Neurosciences and Mental Health, SickKids Research Institute, Toronto, Ontario, Canada
| | - Jamila Aitoubah
- Neurosciences and Mental Health, SickKids Research Institute, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Giovanbattista Grande
- Neurosciences and Mental Health, SickKids Research Institute, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Angela Li
- Neurosciences and Mental Health, SickKids Research Institute, Toronto, Ontario, Canada
| | - Eric W Salter
- Neurosciences and Mental Health, SickKids Research Institute, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Neuroscience, Brown University, Providence, Rhode Island, USA
| | - Alex Wang
- Neurosciences and Mental Health, SickKids Research Institute, Toronto, Ontario, Canada
- Department of Neuroscience, Yale University, New Haven, Connecticut, USA
| | - Melanie D Mark
- Department of Zoology and Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Stefan Herlitze
- Department of Zoology and Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Sean E Egan
- Cell Biology, SickKids Research Institute, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Ontario, Canada
| | - Michael W Salter
- Neurosciences and Mental Health, SickKids Research Institute, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Lu-Yang Wang
- Neurosciences and Mental Health, SickKids Research Institute, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
2
|
Jackson MB, Chiang CW, Cheng J. Fusion pore flux controls the rise-times of quantal synaptic responses. J Gen Physiol 2024; 156:e202313484. [PMID: 38860965 PMCID: PMC11167452 DOI: 10.1085/jgp.202313484] [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: 09/11/2023] [Revised: 03/18/2024] [Accepted: 05/27/2024] [Indexed: 06/12/2024] Open
Abstract
The release of neurotransmitter from a single synaptic vesicle generates a quantal response, which at excitatory synapses in voltage-clamped neurons is referred to as a miniature excitatory postsynaptic current (mEPSC). We analyzed mEPSCs in cultured mouse hippocampal neurons and in HEK cells expressing postsynaptic proteins enabling them to receive synaptic inputs from cocultured neurons. mEPSC amplitudes and rise-times varied widely within and between cells. In neurons, mEPSCs with larger amplitudes had longer rise-times, and this correlation was stronger in neurons with longer mean rise-times. In HEK cells, this correlation was weak and unclear. Standard mechanisms thought to govern mEPSCs cannot account for these results. We therefore developed models to simulate mEPSCs and assess their dependence on different factors. Modeling indicated that longer diffusion times for transmitters released by larger vesicles to reach more distal receptors cannot account for the correlation between rise-time and amplitude. By contrast, incorporating the vesicle size dependence of fusion pore expulsion time recapitulated experimental results well. Larger vesicles produce mEPSCs with larger amplitudes and also take more time to lose their content. Thus, fusion pore flux directly contributes to mEPSC rise-time. Variations in fusion pores account for differences among neurons, between neurons and HEK cells, and the correlation between rise-time and the slope of rise-time versus amplitude plots. Plots of mEPSC amplitude versus rise-time are sensitive to otherwise inaccessible properties of a synapse and offer investigators a means of assessing the role of fusion pores in synaptic release.
Collapse
Affiliation(s)
- Meyer B. Jackson
- Department of Neuroscience, University of Wisconsin—Madison, Madison, WI, USA
| | - Chung-Wei Chiang
- Department of Neuroscience, University of Wisconsin—Madison, Madison, WI, USA
| | - Jinbo Cheng
- Department of Neuroscience, University of Wisconsin—Madison, Madison, WI, USA
| |
Collapse
|
3
|
Scheuer KS, Jansson AM, Zhao X, Jackson MB. Inter and intralaminar excitation of parvalbumin interneurons in mouse barrel cortex. PLoS One 2024; 19:e0289901. [PMID: 38870124 PMCID: PMC11175493 DOI: 10.1371/journal.pone.0289901] [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: 07/27/2023] [Accepted: 04/29/2024] [Indexed: 06/15/2024] Open
Abstract
Parvalbumin (PV) interneurons are inhibitory fast-spiking cells with essential roles in directing the flow of information through cortical circuits. These neurons set the balance between excitation and inhibition and control rhythmic activity. PV interneurons differ between cortical layers in their morphology, circuitry, and function, but how their electrophysiological properties vary has received little attention. Here we investigate responses of PV interneurons in different layers of primary somatosensory barrel cortex (BC) to different excitatory inputs. With the genetically-encoded hybrid voltage sensor, hVOS, we recorded voltage changes in many L2/3 and L4 PV interneurons simultaneously, with stimulation applied to either L2/3 or L4. A semi-automated procedure was developed to identify small regions of interest corresponding to single responsive PV interneurons. Amplitude, half-width, and rise-time were greater for PV interneurons residing in L2/3 compared to L4. Stimulation in L2/3 elicited responses in both L2/3 and L4 with longer latency compared to stimulation in L4. These differences in latency between layers could influence their windows for temporal integration. Thus, PV interneurons in different cortical layers of BC respond in a layer specific and input specific manner, and these differences have potential roles in cortical computations.
Collapse
Affiliation(s)
- Katherine S. Scheuer
- Cellular and Molecular Biology PhD Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Anna M. Jansson
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Xinyu Zhao
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Meyer B. Jackson
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
4
|
Maraslioglu-Sperber A, Pizzi E, Fisch JO, Kattler K, Ritter T, Friauf E. Molecular and functional profiling of cell diversity and identity in the lateral superior olive, an auditory brainstem center with ascending and descending projections. Front Cell Neurosci 2024; 18:1354520. [PMID: 38846638 PMCID: PMC11153811 DOI: 10.3389/fncel.2024.1354520] [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: 12/12/2023] [Accepted: 03/15/2024] [Indexed: 06/09/2024] Open
Abstract
The lateral superior olive (LSO), a prominent integration center in the auditory brainstem, contains a remarkably heterogeneous population of neurons. Ascending neurons, predominantly principal neurons (pLSOs), process interaural level differences for sound localization. Descending neurons (lateral olivocochlear neurons, LOCs) provide feedback into the cochlea and are thought to protect against acoustic overload. The molecular determinants of the neuronal diversity in the LSO are largely unknown. Here, we used patch-seq analysis in mice at postnatal days P10-12 to classify developing LSO neurons according to their functional and molecular profiles. Across the entire sample (n = 86 neurons), genes involved in ATP synthesis were particularly highly expressed, confirming the energy expenditure of auditory neurons. Two clusters were identified, pLSOs and LOCs. They were distinguished by 353 differentially expressed genes (DEGs), most of which were novel for the LSO. Electrophysiological analysis confirmed the transcriptomic clustering. We focused on genes affecting neuronal input-output properties and validated some of them by immunohistochemistry, electrophysiology, and pharmacology. These genes encode proteins such as osteopontin, Kv11.3, and Kvβ3 (pLSO-specific), calcitonin-gene-related peptide (LOC-specific), or Kv7.2 and Kv7.3 (no DEGs). We identified 12 "Super DEGs" and 12 genes showing "Cluster similarity." Collectively, we provide fundamental and comprehensive insights into the molecular composition of individual ascending and descending neurons in the juvenile auditory brainstem and how this may relate to their specific functions, including developmental aspects.
Collapse
Affiliation(s)
- Ayse Maraslioglu-Sperber
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Erika Pizzi
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Jonas O. Fisch
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Kathrin Kattler
- Genetics/Epigenetics Group, Department of Biological Sciences, Saarland University, Saarbrücken, Germany
| | - Tamara Ritter
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Eckhard Friauf
- Animal Physiology Group, Department of Biology, University of Kaiserslautern-Landau, Kaiserslautern, Germany
| |
Collapse
|
5
|
Masumoto N, Kato S, Aichi M, Hasegawa S, Sahara K, Suyama K, Sano A, Miyazaki T, Okudela K, Kaneko T, Takahashi T. AMPAR receptor inhibitors suppress proliferation of human small cell lung cancer cell lines. Thorac Cancer 2023; 14:2897-2908. [PMID: 37605807 PMCID: PMC10569908 DOI: 10.1111/1759-7714.15075] [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: 04/19/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND Small cell lung cancer (SCLC) is a neuroendocrine tumor with poor prognosis. Neuroendocrine tumors possess characteristics of both nerve cells and hormone-secreting cells; therefore, targeting the neuronal properties of these tumors may lead to the development of new therapeutic options. Among the endogenous signaling pathways in the nervous system, targeting the glutamate pathway may be a useful strategy for glioblastoma treatment. Perampanel, an antagonist of the synaptic glutamate α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR), has been reported to be effective in patients with glioblastoma. In this study, we aimed to investigate the antitumor effects of AMPAR antagonists in human SCLC cell lines. METHODS We performed to examine the expression of AMPAR using Western blot and immunohistochemical analysis. The antitumor effects of AMPAR antagonists on human SCLC cell lines were investigated in vitro and in vivo. We also analyzed the signaling pathway of AMPAR antagonists in SCLC cell lines. Statistical analysis was performed by the GraphPad Prism 6 software. RESULTS We first examined the expression of endogenous AMPAR in six human SCLC cell lines, detecting AMPAR proteins in all of them. Next, we tested the anti-proliferative effect of two AMPAR antagonists, talampanel and cyanquixaline, using SCLC cells in vitro and in vivo. Both AMPAR antagonists inhibited cell proliferation and mitogen-activated protein kinase (MAPK) phosphorylation in SCLC cells in vitro. Further, we observed reduced proliferation of implanted cell lines in an in vivo setting, assessed by Ki-67 immunohistochemistry. Additionally, using immunohistochemical analysis we confirmed AMPAR protein expression in human SCLC samples. CONCLUSION AMPAR may be a potential therapeutic target for SCLC.
Collapse
Affiliation(s)
- Nami Masumoto
- Department of PulmonologyYokohama City University Graduate School of MedicineYokohamaJapan
- Department of PhysiologyYokohama City University Graduate School of MedicineYokohamaJapan
- Department of RespirologyNational Hospital Organization Yokohama Medical CenterYokohamaJapan
| | - Shingo Kato
- Department of Clinical Cancer GenomicsYokohama City University HospitalYokohamaJapan
- Department of Gastroenterology and HepatologyYokohama City University Graduate School of MedicineYokohamaJapan
| | - Masahiro Aichi
- Department of PhysiologyYokohama City University Graduate School of MedicineYokohamaJapan
- Department of Obstetrics, Gynecology and Molecular Reproductive ScienceYokohama City University Graduate School of MedicineYokohamaJapan
| | - Sho Hasegawa
- Department of Gastroenterology and HepatologyYokohama City University Graduate School of MedicineYokohamaJapan
| | - Kota Sahara
- Department of PhysiologyYokohama City University Graduate School of MedicineYokohamaJapan
- Department of Gastroenterological SurgeryYokohama City University Graduate School of MedicineYokohamaJapan
| | - Kumiko Suyama
- Department of PhysiologyYokohama City University Graduate School of MedicineYokohamaJapan
| | - Akane Sano
- Department of PhysiologyYokohama City University Graduate School of MedicineYokohamaJapan
| | - Tomoyuki Miyazaki
- Department of PhysiologyYokohama City University Graduate School of MedicineYokohamaJapan
- Center for Promotion of Research and Industry‐Academic Collaboration, Department of Core Project PromotionYokohama City UniversityYokohamaJapan
| | - Koji Okudela
- Department of PathologyYokohama City University Graduate School of MedicineYokohamaJapan
| | - Takeshi Kaneko
- Department of PulmonologyYokohama City University Graduate School of MedicineYokohamaJapan
| | - Takuya Takahashi
- Department of PhysiologyYokohama City University Graduate School of MedicineYokohamaJapan
| |
Collapse
|
6
|
Scheuer KS, Jansson AM, Zhao X, Jackson MB. Inter and Intralaminar Excitation of Parvalbumin Interneurons in Mouse Barrel Cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.02.543448. [PMID: 37398428 PMCID: PMC10312540 DOI: 10.1101/2023.06.02.543448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Parvalbumin (PV) interneurons are inhibitory fast-spiking cells with essential roles in directing the flow of information through cortical circuits. These neurons set the balance between excitation and inhibition, control rhythmic activity, and have been linked to disorders including autism spectrum and schizophrenia. PV interneurons differ between cortical layers in their morphology, circuitry, and function, but how their electrophysiological properties vary has received little attention. Here we investigate responses of PV interneurons in different layers of primary somatosensory barrel cortex (BC) to different excitatory inputs. With the genetically-encoded hybrid voltage sensor, hVOS, we recorded voltage changes simultaneously in many L2/3 and L4 PV interneurons to stimulation in either L2/3 or L4. Decay-times were consistent across L2/3 and L4. Amplitude, half-width, and rise-time were greater for PV interneurons residing in L2/3 compared to L4. Stimulation in L2/3 elicited responses in both L2/3 and L4 with longer latency compared to stimulation in L4. These differences in latency between layers could influence their windows for temporal integration. Thus PV interneurons in different cortical layers of BC show differences in response properties with potential roles in cortical computations.
Collapse
Affiliation(s)
- Kate S Scheuer
- Cellular and Molecular Biology Program, University of Wisconsin-Madison, Madison, Wisconsin, 53705
| | - Anna M Jansson
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, 53705
| | - Xinyu Zhao
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, 53705
| | - Meyer B Jackson
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin, 53705
| |
Collapse
|
7
|
Rutherford MA, Bhattacharyya A, Xiao M, Cai HM, Pal I, Rubio ME. GluA3 subunits are required for appropriate assembly of AMPAR GluA2 and GluA4 subunits on cochlear afferent synapses and for presynaptic ribbon modiolar-pillar morphology. eLife 2023; 12:e80950. [PMID: 36648432 PMCID: PMC9891727 DOI: 10.7554/elife.80950] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 01/16/2023] [Indexed: 01/18/2023] Open
Abstract
Cochlear sound encoding depends on α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (AMPARs), but reliance on specific pore-forming subunits is unknown. With 5-week-old male C57BL/6J Gria3-knockout mice (i.e., subunit GluA3KO) we determined cochlear function, synapse ultrastructure, and AMPAR molecular anatomy at ribbon synapses between inner hair cells (IHCs) and spiral ganglion neurons. GluA3KO and wild-type (GluA3WT) mice reared in ambient sound pressure level (SPL) of 55-75 dB had similar auditory brainstem response (ABR) thresholds, wave-1 amplitudes, and latencies. Postsynaptic densities (PSDs), presynaptic ribbons, and synaptic vesicle sizes were all larger on the modiolar side of the IHCs from GluA3WT, but not GluA3KO, demonstrating GluA3 is required for modiolar-pillar synapse differentiation. Presynaptic ribbons juxtaposed with postsynaptic GluA2/4 subunits were similar in quantity, however, lone ribbons were more frequent in GluA3KO and GluA2-lacking synapses were observed only in GluA3KO. GluA2 and GluA4 immunofluorescence volumes were smaller on the pillar side than the modiolar side in GluA3KO, despite increased pillar-side PSD size. Overall, the fluorescent puncta volumes of GluA2 and GluA4 were smaller in GluA3KO than GluA3WT. However, GluA3KO contained less GluA2 and greater GluA4 immunofluorescence intensity relative to GluA3WT (threefold greater mean GluA4:GluA2 ratio). Thus, GluA3 is essential in development, as germline disruption of Gria3 caused anatomical synapse pathology before cochlear output became symptomatic by ABR. We propose the hearing loss in older male GluA3KO mice results from progressive synaptopathy evident in 5-week-old mice as decreased abundance of GluA2 subunits and an increase in GluA2-lacking, GluA4-monomeric Ca2+-permeable AMPARs.
Collapse
Affiliation(s)
- Mark A Rutherford
- Department of Otolaryngology, Washington University School of MedicineSt LouisUnited States
| | - Atri Bhattacharyya
- Department of Otolaryngology, Washington University School of MedicineSt LouisUnited States
| | - Maolei Xiao
- Department of Otolaryngology, Washington University School of MedicineSt LouisUnited States
| | - Hou-Ming Cai
- Department of Neurobiology, University of Pittsburgh School of MedicinePittsburghUnited States
| | - Indra Pal
- Department of Neurobiology, University of Pittsburgh School of MedicinePittsburghUnited States
| | - Maria Eulalia Rubio
- Department of Neurobiology, University of Pittsburgh School of MedicinePittsburghUnited States
- Department of Otolaryngology, University of Pittsburgh School of MedicinePittsburghUnited States
| |
Collapse
|
8
|
Han Y, Cao R, Qin L, Chen LY, Tang AH, Südhof TC, Zhang B. Neuroligin-3 confines AMPA receptors into nanoclusters, thereby controlling synaptic strength at the calyx of Held synapses. SCIENCE ADVANCES 2022; 8:eabo4173. [PMID: 35704570 PMCID: PMC9200272 DOI: 10.1126/sciadv.abo4173] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 05/02/2022] [Indexed: 05/30/2023]
Abstract
The subsynaptic organization of postsynaptic neurotransmitter receptors into nanoclusters that are aligned with presynaptic release sites is essential for the high fidelity of synaptic transmission. However, the mechanisms controlling the nanoscale organization of neurotransmitter receptors in vivo remain incompletely understood. Here, we deconstructed the role of neuroligin-3 (Nlgn3), a postsynaptic adhesion molecule linked to autism, in organizing AMPA-type glutamate receptors in the calyx of Held synapse. Deletion of Nlgn3 lowered the amplitude and slowed the kinetics of AMPA receptor-mediated synaptic responses. Super-resolution microscopy revealed that, unexpectedly, these impairments in synaptic transmission were associated with an increase in the size of postsynaptic PSD-95 and AMPA receptor nanoclusters but a decrease of the densities in these clusters. Modeling showed that a dilution of AMPA receptors into larger nanocluster volumes decreases synaptic strength. Nlgn3, likely by binding to presynaptic neurexins, thus is a key organizer of AMPA receptor nanoclusters that likely acts via PSD-95 adaptors to optimize the fidelity of synaptic transmission.
Collapse
Affiliation(s)
- Ying Han
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Ran Cao
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230026, China
- CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles & Cellular Dynamics and Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Liming Qin
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Lulu Y. Chen
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA 92697, USA
- Department of Molecular and Cellular Physiology, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94043, USA
| | - Ai-Hui Tang
- Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230026, China
- CAS Key Laboratory of Brain Function and Disease, Ministry of Education Key Laboratory for Membrane-less Organelles & Cellular Dynamics and Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Thomas C. Südhof
- Department of Molecular and Cellular Physiology, Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94043, USA
| | - Bo Zhang
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen 518132, China
| |
Collapse
|
9
|
Richardson A, Ciampani V, Stancu M, Bondarenko K, Newton S, Steinert JR, Pilati N, Graham BP, Kopp-Scheinpflug C, Forsythe ID. Kv3.3 subunits control presynaptic action potential waveform and neurotransmitter release at a central excitatory synapse. eLife 2022; 11:75219. [PMID: 35510987 PMCID: PMC9110028 DOI: 10.7554/elife.75219] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 04/29/2022] [Indexed: 11/29/2022] Open
Abstract
Kv3 potassium currents mediate rapid repolarisation of action potentials (APs), supporting fast spikes and high repetition rates. Of the four Kv3 gene family members, Kv3.1 and Kv3.3 are highly expressed in the auditory brainstem and we exploited this to test for subunit-specific roles at the calyx of Held presynaptic terminal in the mouse. Deletion of Kv3.3 (but not Kv3.1) reduced presynaptic Kv3 channel immunolabelling, increased presynaptic AP duration and facilitated excitatory transmitter release; which in turn enhanced short-term depression during high-frequency transmission. The response to sound was delayed in the Kv3.3KO, with higher spontaneous and lower evoked firing, thereby reducing signal-to-noise ratio. Computational modelling showed that the enhanced EPSC and short-term depression in the Kv3.3KO reflected increased vesicle release probability and accelerated activity-dependent vesicle replenishment. We conclude that Kv3.3 mediates fast repolarisation for short precise APs, conserving transmission during sustained high-frequency activity at this glutamatergic excitatory synapse.
Collapse
Affiliation(s)
- Amy Richardson
- epartment of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, United Kingdom
| | - Victoria Ciampani
- epartment of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, United Kingdom
| | - Mihai Stancu
- Division of Neurobiology, Ludwig-Maximilians-Universität München, Munchen, Germany
| | - Kseniia Bondarenko
- epartment of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, United Kingdom
| | - Sherylanne Newton
- epartment of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, United Kingdom
| | - Joern R Steinert
- epartment of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, United Kingdom
| | - Nadia Pilati
- Istituto di Ricerca Pediatrica Citta'della Speranza, Padova, Italy
| | - Bruce P Graham
- Computing Science and Mathematics, University of Stirling, Stirling, United Kingdom
| | | | - Ian D Forsythe
- epartment of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
10
|
Impaired Subcortical Processing of Amplitude-Modulated Tones in Mice Deficient for Cacna2d3, a Risk Gene for Autism Spectrum Disorders in Humans. eNeuro 2022; 9:ENEURO.0118-22.2022. [PMID: 35410870 PMCID: PMC9034753 DOI: 10.1523/eneuro.0118-22.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 03/21/2022] [Indexed: 12/18/2022] Open
Abstract
Temporal processing of complex sounds is a fundamental and complex task in hearing and a prerequisite for processing and understanding vocalization, speech, and prosody. Here, we studied response properties of neurons in the inferior colliculus (IC) in mice lacking Cacna2d3, a risk gene for autism spectrum disorders (ASDs). The α2δ3 auxiliary Ca2+ channel subunit encoded by Cacna2d3 is essential for proper function of glutamatergic synapses in the auditory brainstem. Recent evidence has shown that much of auditory feature extraction is performed in the auditory brainstem and IC, including processing of amplitude modulation (AM). We determined both spectral and temporal properties of single- and multi-unit responses in the IC of anesthetized mice. IC units of α2δ3−/− mice showed normal tuning properties yet increased spontaneous rates compared with α2δ3+/+. When stimulated with AM tones, α2δ3−/− units exhibited less precise temporal coding and reduced evoked rates to higher modulation frequencies (fm). Whereas first spike latencies (FSLs) were increased for only few modulation frequencies, population peak latencies were increased for fm ranging from 20 to 100 Hz in α2δ3−/− IC units. The loss of precision of temporal coding with increasing fm from 70 to 160 Hz was characterized using a normalized offset-corrected (Pearson-like) correlation coefficient, which appeared more appropriate than the metrics of vector strength. The processing deficits of AM sounds analyzed at the level of the IC indicate that α2δ3−/− mice exhibit a subcortical auditory processing disorder (APD). Similar deficits may be present in other mouse models for ASDs.
Collapse
|
11
|
García-Hernández S, Rubio ME. Role of GluA4 in the acoustic and tactile startle responses. Hear Res 2022; 414:108410. [PMID: 34915397 PMCID: PMC8776314 DOI: 10.1016/j.heares.2021.108410] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 08/26/2021] [Accepted: 12/06/2021] [Indexed: 02/03/2023]
Abstract
The primary startle response (SR) is an innate reaction evoked by sudden and intense acoustic, tactile or visual stimuli. In rodents and humans the SR involves reflexive contractions of the face, neck and limb muscles. The acoustic startle response (ASR) pathway consists of auditory nerve fibers (AN), cochlear root neurons (CRNs) and giant neurons of the caudal pontine reticular nucleus (PnC), which synapse on cranial and spinal motor neurons. The tactile startle response (TSR) is transmitted by primary sensory neurons to the principal sensory (Pr5) and spinal (Sp5) trigeminal nuclei. The ventral part of Pr5 projects directly to the PnC neurons. The SR requires rapid transmission of sensory information to initiate a fast motor response. Alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPAR) are necessary to transmit auditory information to the PnC neurons and elicit the SR. AMPARs containing the glutamate AMPAR subunit 4 (GluA4) have fast kinetics, which makes them ideal candidates to transmit the SR signal. This study examined the role of GluA4 within the primary SR pathway by using GluA4 knockout (GluA4-KO) mice. Deletion of GluA4 considerably decreased the amplitude and probability of successful ASR and TSR, indicating that the presence of this subunit is critical at a common station within the startle pathway. We conclude that deletion of GluA4 affects the transmission of sensory signals from acoustic and tactile pathways to the motor component of the startle reflex. Therefore, GluA4 is required for the full response and for reliable elicitation of the startle response.
Collapse
Affiliation(s)
- Sofía García-Hernández
- Corresponding authors. Sofía García-Hernández, María E. Rubio, Departments of Neurobiology and Otolaryngology, University of Pittsburgh Medical School, BST3 Building, 3501 Fifth Avenue #10016, Pittsburgh, PA 15261, ,
| | - María E. Rubio
- Corresponding authors. Sofía García-Hernández, María E. Rubio, Departments of Neurobiology and Otolaryngology, University of Pittsburgh Medical School, BST3 Building, 3501 Fifth Avenue #10016, Pittsburgh, PA 15261, ,
| |
Collapse
|
12
|
Krohs C, Körber C, Ebbers L, Altaf F, Hollje G, Hoppe S, Dörflinger Y, Prosser HM, Nothwang HG. Loss of miR-183/96 Alters Synaptic Strength via Presynaptic and Postsynaptic Mechanisms at a Central Synapse. J Neurosci 2021; 41:6796-6811. [PMID: 34193555 PMCID: PMC8360680 DOI: 10.1523/jneurosci.0139-20.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/03/2021] [Accepted: 05/09/2021] [Indexed: 12/27/2022] Open
Abstract
A point mutation in miR-96 causes non-syndromic progressive peripheral hearing loss and alters structure and physiology of the central auditory system. To gain further insight into the functions of microRNAs (miRNAs) within the central auditory system, we investigated constitutive Mir-183/96dko mice of both sexes. In this mouse model, the genomically clustered miR-183 and miR-96 are constitutively deleted. It shows significantly and specifically reduced volumes of auditory hindbrain nuclei, because of decreases in cell number and soma size. Electrophysiological analysis of the calyx of Held synapse in the medial nucleus of the trapezoid body (MNTB) demonstrated strongly altered synaptic transmission in young-adult mice. We observed an increase in quantal content and readily releasable vesicle pool size in the presynapse while the overall morphology of the calyx was unchanged. Detailed analysis of the active zones (AZs) revealed differences in its molecular composition and synaptic vesicle (SV) distribution. Postsynaptically, altered clustering and increased synaptic abundancy of the AMPA receptor subunit GluA1 was observed resulting in an increase in quantal amplitude. Together, these presynaptic and postsynaptic alterations led to a 2-fold increase of the evoked excitatory postsynaptic currents in MNTB neurons. None of these changes were observed in deaf Cldn14ko mice, confirming an on-site role of miR-183 and miR-96 in the auditory hindbrain. Our data suggest that the Mir-183/96 cluster plays a key role for proper synaptic transmission at the calyx of Held and for the development of the auditory hindbrain.SIGNIFICANCE STATEMENT The calyx of Held is the outstanding model system to study basic synaptic physiology. Yet, genetic factors driving its morphologic and functional maturation are largely unknown. Here, we identify the Mir-183/96 cluster as an important factor to regulate its synaptic strength. Presynaptically, Mir-183/96dko calyces show an increase in release-ready synaptic vesicles (SVs), quantal content and abundance of the proteins Bassoon and Piccolo. Postsynaptically, the quantal size as well as number and size of GluA1 puncta were increased. The two microRNAs (miRNAs) are thus attractive candidates for regulation of synaptic maturation and long-term adaptations to sound levels. Moreover, the different phenotypic outcomes of different types of mutations in the Mir-183 cluster corroborate the requirement of mutation-tailored therapies in patients with hearing loss.
Collapse
Affiliation(s)
- Constanze Krohs
- Division of Neurogenetics, Department of Neuroscience, Carl von Ossietzky University Oldenburg, Oldenburg 26129, Germany
| | - Christoph Körber
- Institute of Anatomy und Cell Biology, Department of Functional Neuroanatomy, Heidelberg University, Heidelberg 69120, Germany
| | - Lena Ebbers
- Division of Neurogenetics, Department of Neuroscience, Carl von Ossietzky University Oldenburg, Oldenburg 26129, Germany
| | - Faiza Altaf
- Division of Neurogenetics, Department of Neuroscience, Carl von Ossietzky University Oldenburg, Oldenburg 26129, Germany
| | - Giulia Hollje
- Division of Neurogenetics, Department of Neuroscience, Carl von Ossietzky University Oldenburg, Oldenburg 26129, Germany
| | - Simone Hoppe
- Institute of Anatomy und Cell Biology, Department of Functional Neuroanatomy, Heidelberg University, Heidelberg 69120, Germany
| | - Yvette Dörflinger
- Institute of Anatomy und Cell Biology, Department of Functional Neuroanatomy, Heidelberg University, Heidelberg 69120, Germany
| | - Haydn M Prosser
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Cambridge CB10 1SA, United Kingdom
| | - Hans Gerd Nothwang
- Division of Neurogenetics, Department of Neuroscience, Carl von Ossietzky University Oldenburg, Oldenburg 26129, Germany
- Excellence Cluster Hearing4all, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
| |
Collapse
|
13
|
Baranovic J. AMPA receptors in the synapse: Very little space and even less time. Neuropharmacology 2021; 196:108711. [PMID: 34271021 DOI: 10.1016/j.neuropharm.2021.108711] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 06/30/2021] [Accepted: 07/08/2021] [Indexed: 12/14/2022]
Abstract
Glutamate is by far the most abundant neurotransmitter used by excitatory synapses in the vertebrate central nervous system. Once released into the synaptic cleft, it depolarises the postsynaptic membrane and activates downstream signalling pathways resulting in the propagation of the excitatory signal. Initial depolarisation is primarily mediated by α-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA) receptors. These ion channels are the first ones to be activated by released glutamate and their kinetics, dynamics and abundance on the postsynaptic membrane defines the strength of the postsynaptic response. This review focuses on native AMPA receptors and synaptic environment they inhabit and considers structural and functional properties of the receptors obtained in heterologous systems in the light of spatial and temporal constraints of the synapse. This article is part of the special Issue on 'Glutamate Receptors - AMPA receptors'.
Collapse
Affiliation(s)
- Jelena Baranovic
- School of Biological Sciences, University of Edinburgh, King's Buildings, Max Born Crescent, EH9 3BF, Edinburgh, UK.
| |
Collapse
|
14
|
Gurma M, Yang YM, Wang LY. Developmental plasticity of NMDA receptors at the calyx of Held synapse. Neuropharmacology 2021; 196:108697. [PMID: 34242682 DOI: 10.1016/j.neuropharm.2021.108697] [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: 02/15/2021] [Revised: 06/11/2021] [Accepted: 06/12/2021] [Indexed: 11/25/2022]
Abstract
Excitatory synaptic transmission is largely mediated by glutamate receptors in central synapses, such as the calyx of Held synapse in the auditory brainstem. This synapse is best known for undergoing extensive morphological and functional changes throughout early development and thereby has served as a prominent model system to study presynaptic mechanisms of neurotransmitter release. However, the pivotal roles of N-methyl-d-aspartate receptors (NMDARs) in gating acute forms of activity-dependent, persistent plasticity in vitro and chronic developmental remodeling in vivo are hardly noted. This article will provide a retrospective review of key experimental evidence to conceptualize the impact of a transient abundance of NMDARs during the early postnatal stage on the functionality of fast-spiking central synapses while raising a series of outstanding questions that are of general significance for understanding the developing brain in health and diseases. This article is part of the special Issue on "Glutamate Receptors - NMDA receptors".
Collapse
Affiliation(s)
- Maria Gurma
- Program in Neurosciences & Mental Health, SickKids Research Institute, 555 University Ave, Toronto, Ontario M5G 1X8, Canada; Department of Physiology, University of Toronto, 1 Kings Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Yi-Mei Yang
- Department of Biomedical Sciences, University of Minnesota, Duluth MN, 55812, USA
| | - Lu-Yang Wang
- Program in Neurosciences & Mental Health, SickKids Research Institute, 555 University Ave, Toronto, Ontario M5G 1X8, Canada; Department of Physiology, University of Toronto, 1 Kings Circle, Toronto, Ontario, M5S 1A8, Canada.
| |
Collapse
|
15
|
Walia A, Lee C, Hartsock J, Goodman SS, Dolle R, Salt AN, Lichtenhan JT, Rutherford MA. Reducing Auditory Nerve Excitability by Acute Antagonism of Ca 2+-Permeable AMPA Receptors. Front Synaptic Neurosci 2021; 13:680621. [PMID: 34290596 PMCID: PMC8287724 DOI: 10.3389/fnsyn.2021.680621] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 05/31/2021] [Indexed: 11/13/2022] Open
Abstract
Hearing depends on glutamatergic synaptic transmission mediated by α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs). AMPARs are tetramers, where inclusion of the GluA2 subunit reduces overall channel conductance and Ca2+ permeability. Cochlear afferent synapses between inner hair cells (IHCs) and auditory nerve fibers (ANFs) contain the AMPAR subunits GluA2, 3, and 4. However, the tetrameric complement of cochlear AMPAR subunits is not known. It was recently shown in mice that chronic intracochlear delivery of IEM-1460, an antagonist selective for GluA2-lacking AMPARs [also known as Ca2+-permeable AMPARs (CP-AMPARs)], before, during, and after acoustic overexposure prevented both the trauma to ANF synapses and the ensuing reduction of cochlear nerve activity in response to sound. Surprisingly, baseline measurements of cochlear function before exposure were unaffected by chronic intracochlear delivery of IEM-1460. This suggested that cochlear afferent synapses contain GluA2-lacking CP-AMPARs alongside GluA2-containing Ca2+-impermeable AMPA receptors (CI-AMPARs), and that the former can be antagonized for protection while the latter remain conductive. Here, we investigated hearing function in the guinea pig during acute local or systemic delivery of CP-AMPAR antagonists. Acute intracochlear delivery of IEM-1460 or systemic delivery of IEM-1460 or IEM-1925 reduced the amplitude of the ANF compound action potential (CAP) significantly, for all tone levels and frequencies, by > 50% without affecting CAP thresholds or distortion product otoacoustic emissions (DPOAE). Following systemic dosing, IEM-1460 levels in cochlear perilymph were ~ 30% of blood levels, on average, consistent with pharmacokinetic properties predicting permeation of the compounds into the brain and ear. Both compounds were metabolically stable with half-lives >5 h in vitro, and elimination half-lives in vivo of 118 min (IEM-1460) and 68 min (IEM-1925). Heart rate monitoring and off-target binding assays suggest an enhanced safety profile for IEM-1925 over IEM-1460. Compound potency on CAP reduction (IC50 ~ 73 μM IEM-1460) was consistent with a mixture of GluA2-lacking and GluA2-containing AMPARs. These data strongly imply that cochlear afferent synapses of the guinea pig contain GluA2-lacking CP-AMPARs. We propose these CP-AMPARs may be acutely antagonized with systemic dosing, to protect from glutamate excitotoxicity, while transmission at GluA2-containing AMPARs persists to mediate hearing during the protection.
Collapse
Affiliation(s)
- Amit Walia
- Department of Otolaryngology, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Choongheon Lee
- Department of Otolaryngology, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Jared Hartsock
- Department of Otolaryngology, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Shawn S Goodman
- Department of Communication Sciences and Disorders, University of Iowa, Iowa City, IA, United States
| | - Roland Dolle
- Department of Biochemistry and Molecular Biophysics, Washington University Center for Drug Discovery, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Alec N Salt
- Department of Otolaryngology, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Jeffery T Lichtenhan
- Department of Otolaryngology, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Mark A Rutherford
- Department of Otolaryngology, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| |
Collapse
|
16
|
Kita K, Albergaria C, Machado AS, Carey MR, Müller M, Delvendahl I. GluA4 facilitates cerebellar expansion coding and enables associative memory formation. eLife 2021; 10:65152. [PMID: 34219651 PMCID: PMC8291978 DOI: 10.7554/elife.65152] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 07/01/2021] [Indexed: 01/17/2023] Open
Abstract
AMPA receptors (AMPARs) mediate excitatory neurotransmission in the central nervous system (CNS) and their subunit composition determines synaptic efficacy. Whereas AMPAR subunits GluA1–GluA3 have been linked to particular forms of synaptic plasticity and learning, the functional role of GluA4 remains elusive. Here, we demonstrate a crucial function of GluA4 for synaptic excitation and associative memory formation in the cerebellum. Notably, GluA4-knockout mice had ~80% reduced mossy fiber to granule cell synaptic transmission. The fidelity of granule cell spike output was markedly decreased despite attenuated tonic inhibition and increased NMDA receptor-mediated transmission. Computational network modeling incorporating these changes revealed that deletion of GluA4 impairs granule cell expansion coding, which is important for pattern separation and associative learning. On a behavioral level, while locomotor coordination was generally spared, GluA4-knockout mice failed to form associative memories during delay eyeblink conditioning. These results demonstrate an essential role for GluA4-containing AMPARs in cerebellar information processing and associative learning.
Collapse
Affiliation(s)
- Katarzyna Kita
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, Zurich, Switzerland
| | - Catarina Albergaria
- Champalimaud Neuroscience Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Ana S Machado
- Champalimaud Neuroscience Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Megan R Carey
- Champalimaud Neuroscience Programme, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Martin Müller
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, Zurich, Switzerland
| | - Igor Delvendahl
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, Zurich, Switzerland
| |
Collapse
|
17
|
Zemaitis K, Kaliyappan K, Frerichs V, Friedman A, Krishnan Muthaiah VP. Mass spectrometry imaging of blast overpressure induced modulation of GABA/glutamate levels in the central auditory neuraxis of Chinchilla. Exp Mol Pathol 2021; 119:104605. [PMID: 33453279 PMCID: PMC11620843 DOI: 10.1016/j.yexmp.2021.104605] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 01/09/2021] [Accepted: 01/10/2021] [Indexed: 11/15/2022]
Abstract
Acoustic trauma damages inner ear neural structures including cochlear hair cells which result in hearing loss and neurotransmitter imbalances within the synapses of the central auditory pathway. Disruption of GABA/glutamate levels underlies, tinnitus, a phantom perception of sound that persists post-exposure to blast noise which may manifest in tandem with acute/chronic loss of hearing. Many putative theories explain tinnitus physiology based on indirect and direct assays in animal models and humans, although there is no comprehensive evidence to explain the phenomenon. Here, GABA/glutamate levels were imaged and quantified in a blast overpressure model of chinchillas using Fourier transform ion cyclotron resonance mass spectrometry imaging. The direct measurement from whole-brain sections identified the relative levels of GABA/glutamate in the central auditory neuraxis centers including the cochlear nucleus, inferior colliculus, and auditory cortex. These preliminary results provide insight on the homeostasis of GABA/glutamate within whole-brain sections of chinchilla for investigation of the pathomechanism of blast-induced tinnitus.
Collapse
Affiliation(s)
- Kevin Zemaitis
- Chemistry Instrument Center, Department of Chemistry, Natural Sciences Complex, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | - Kathiravan Kaliyappan
- Department of Rehabilitation Sciences, School of Publich Health and Health Professions, Kimball Tower, University at Buffalo, State University of New York, Buffalo, NY 14215, USA
| | - Valerie Frerichs
- Chemistry Instrument Center, Department of Chemistry, Natural Sciences Complex, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | - Alan Friedman
- Department of Materials Design and Innovation, School of Engineering and Applied Sciences, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | - Vijaya Prakash Krishnan Muthaiah
- Department of Rehabilitation Sciences, School of Publich Health and Health Professions, Kimball Tower, University at Buffalo, State University of New York, Buffalo, NY 14215, USA.
| |
Collapse
|
18
|
Abstract
During development and adulthood, the normal activity of the auditory nerve plays a critical role in the maintenance of both fundamental structural, molecular, and functional parameters of auditory nerve synapses, and the postsynaptic excitatory or inhibitory neurons within the cochlear nucleus (CN). In addition, normal activity within the synaptic circuits of the CN is key to developing and maintaining appropriate synapse connectivity as well as the initiation of binaural sound processing in the superior olivary complex (SOC). Development plays a critical role in the proper neuronal connectivity and establishes a topographic map along the entire auditory pathway. Furthermore, evidence shows that neurons and synaptic circuits in the auditory brainstem are not hard-wired, but instead are plastic in response to hearing deficits. Whether this plasticity in response to hearing loss is compensatory or pathological is still unknown.
Collapse
Affiliation(s)
- María Eulalia Rubio
- Departments of Neurobiology and Otolaryngology, University of Pittsburgh, School of Medicine, BST3 Building, room #10016, 3501 Fifth Venue, Pittsburgh, PA, 15261
| |
Collapse
|
19
|
Role of GluA3 AMPA Receptor Subunits in the Presynaptic and Postsynaptic Maturation of Synaptic Transmission and Plasticity of Endbulb-Bushy Cell Synapses in the Cochlear Nucleus. J Neurosci 2020; 40:2471-2484. [PMID: 32051325 DOI: 10.1523/jneurosci.2573-19.2020] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/22/2020] [Accepted: 02/02/2020] [Indexed: 11/21/2022] Open
Abstract
The AMPA receptor (AMPAR) subunit GluA3 has been suggested to shape synaptic transmission and activity-dependent plasticity in endbulb-bushy cell synapses (endbulb synapses) in the anteroventral cochlear nucleus, yet the specific roles of GluA3 in the synaptic transmission at endbulb synapses remains unexplored. Here, we compared WT and GluA3 KO mice of both sexes and identified several important roles of GluA3 in the maturation of synaptic transmission and short-term plasticity in endbulb synapses. We show that GluA3 largely determines the ultrafast kinetics of endbulb synapses glutamatergic currents by promoting the insertion of postsynaptic AMPARs that contain fast desensitizing flop subunits. In addition, GluA3 is also required for the normal function, structure, and development of the presynaptic terminal which leads to altered short term-depression in GluA3 KO mice. The presence of GluA3 reduces and slows synaptic depression, which is achieved by lowering the probability of vesicle release, promoting efficient vesicle replenishment, and increasing the readily releasable pool of synaptic vesicles. Surprisingly, GluA3 also makes the speed of synaptic depression rate-invariant. We propose that the slower and rate-invariant speed of depression allows an initial response window that still contains presynaptic firing rate information before the synapse is depressed. Because this response window is rate-invariant, GluA3 extends the range of presynaptic firing rates over which rate information in bushy cells can be preserved. This novel role of GluA3 may be important to allowing the postsynaptic targets of spherical bushy cells in mice use rate information for encoding sound intensity and sound localization.SIGNIFICANCE STATEMENT We report novel roles of the glutamate receptor subunit GluA3 in synaptic transmission in synapses between auditory nerve fibers and spherical bushy cells (BCs) in the cochlear nucleus. We show that GluA3 contributes to the generation of ultrafast glutamatergic currents at these synapses, which is important to preserve temporal information about the sound. Furthermore, we demonstrate that GluA3 contributes to the normal function and development of the presynaptic terminal, whose properties shape short-term plasticity. GluA3 slows and attenuates synaptic depression, and makes it less dependent on the presynaptic firing rates. This may help BCs to transfer information about the high rates of activity that occur at the synapse in vivo to postsynaptic targets that use rate information for sound localization.
Collapse
|
20
|
Lesperance LS, Yang YM, Wang LY. Delayed expression of activity-dependent gating switch in synaptic AMPARs at a central synapse. Mol Brain 2020; 13:6. [PMID: 31941524 PMCID: PMC6961283 DOI: 10.1186/s13041-019-0536-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 12/17/2019] [Indexed: 01/11/2023] Open
Abstract
Developing central synapses exhibit robust plasticity and undergo experience-dependent remodeling. Evidently, synapses in sensory systems such as auditory brainstem circuits mature rapidly to achieve high-fidelity neurotransmission for sound localization. This depends on a developmental switch in AMPAR composition from slow-gating GluA1-dominant to fast-gating GluA4-dominant, but the mechanisms underlying this switch remain unknown. We hypothesize that patterned stimuli mimicking spontaneous/sound evoked activity in the early postnatal stage drives this gating switch. We examined activity-dependent changes in evoked and miniature excitatory postsynaptic currents (eEPSCs and mEPSCs) at the calyx of Held synapse by breaking through the postsynaptic membrane at different time points following 2 min of theta burst stimulation (TBS) to afferents in mouse brainstem slices. We found the decay time course of eEPSCs accelerated, but this change was not apparent until > 30 min after TBS. Histogram analyses of the decay time constants of mEPSCs for naive and tetanized synapses revealed two populations centered around τfast ≈ 0.4 and 0.8 ms, but the relative weight of the τ0.4 population over the τ0.8 population increased significantly only in tetanized synapses. Such changes are blocked by NMDAR or mGluR1/5 antagonists or inhibitors of CaMKII, PKC and protein synthesis, and more importantly precluded in GluA4−/− synapses, suggesting GluA4 is the substrate underlying the acceleration. Our results demonstrate a novel form of plasticity working through NMDAR and mGluR activation to trigger a gating switch of AMPARs with a temporally delayed onset of expression, ultimately enhancing the development of high-fidelity synaptic transmission.
Collapse
Affiliation(s)
- Lee Stephen Lesperance
- Program in Neurosciences & Mental Health, SickKids Research Institute, 555 University Ave, Toronto, Ontario, M5G 1X8, Canada
| | - Yi-Mei Yang
- Program in Neurosciences & Mental Health, SickKids Research Institute, 555 University Ave, Toronto, Ontario, M5G 1X8, Canada.,Department of Biomedical Sciences, University of Minnesota, Duluth, MN 55812, USA
| | - Lu-Yang Wang
- Program in Neurosciences & Mental Health, SickKids Research Institute, 555 University Ave, Toronto, Ontario, M5G 1X8, Canada.
| |
Collapse
|
21
|
Rapid and sustained homeostatic control of presynaptic exocytosis at a central synapse. Proc Natl Acad Sci U S A 2019; 116:23783-23789. [PMID: 31685637 PMCID: PMC6876255 DOI: 10.1073/pnas.1909675116] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Homeostatic mechanisms stabilize neural activity, and there are genetic links between homeostatic plasticity and neural disease. While homeostatic plasticity in the central nervous system (CNS) operates on relatively slow time scales of hours to days, activity-dependent forms of synaptic plasticity alter neural activity on much faster time scales. It is unclear if homeostatic plasticity stabilizes CNS synapses on rapid time scales. Here, we uncovered that cerebellar synapses stabilize transmission within minutes upon activity perturbation. This is achieved through homeostatic control of presynaptic exocytosis. We show that synergistic modulation of distinct presynaptic mechanisms not only maintains synaptic efficacy on rapid, but also on prolonged time scales. Homeostatic control of presynaptic exocytosis may be a general mechanism for stabilizing CNS function. Animal behavior is remarkably robust despite constant changes in neural activity. Homeostatic plasticity stabilizes central nervous system (CNS) function on time scales of hours to days. If and how CNS function is stabilized on more rapid time scales remains unknown. Here, we discovered that mossy fiber synapses in the mouse cerebellum homeostatically control synaptic efficacy within minutes after pharmacological glutamate receptor impairment. This rapid form of homeostatic plasticity is expressed presynaptically. We show that modulations of readily releasable vesicle pool size and release probability normalize synaptic strength in a hierarchical fashion upon acute pharmacological and prolonged genetic receptor perturbation. Presynaptic membrane capacitance measurements directly demonstrate regulation of vesicle pool size upon receptor impairment. Moreover, presynaptic voltage-clamp analysis revealed increased Ca2+-current density under specific experimental conditions. Thus, homeostatic modulation of presynaptic exocytosis through specific mechanisms stabilizes synaptic transmission in a CNS circuit on time scales ranging from minutes to months. Rapid presynaptic homeostatic plasticity may ensure stable neural circuit function in light of rapid activity-dependent plasticity.
Collapse
|
22
|
Frank CA, James TD, Müller M. Homeostatic control of Drosophila neuromuscular junction function. Synapse 2019; 74:e22133. [PMID: 31556149 PMCID: PMC6817395 DOI: 10.1002/syn.22133] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/05/2019] [Accepted: 09/19/2019] [Indexed: 02/06/2023]
Abstract
The ability to adapt to changing internal and external conditions is a key feature of biological systems. Homeostasis refers to a regulatory process that stabilizes dynamic systems to counteract perturbations. In the nervous system, homeostatic mechanisms control neuronal excitability, neurotransmitter release, neurotransmitter receptors, and neural circuit function. The neuromuscular junction (NMJ) of Drosophila melanogaster has provided a wealth of molecular information about how synapses implement homeostatic forms of synaptic plasticity, with a focus on the transsynaptic, homeostatic modulation of neurotransmitter release. This review examines some of the recent findings from the Drosophila NMJ and highlights questions the field will ponder in coming years.
Collapse
Affiliation(s)
- C Andrew Frank
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Interdisciplinary Programs in Neuroscience, Genetics, and Molecular Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Thomas D James
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, Iowa, USA
| | - Martin Müller
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland.,Neuroscience Center Zurich, Zurich, Switzerland
| |
Collapse
|
23
|
Lujan B, Dagostin A, von Gersdorff H. Presynaptic Diversity Revealed by Ca 2+-Permeable AMPA Receptors at the Calyx of Held Synapse. J Neurosci 2019; 39:2981-2994. [PMID: 30679394 PMCID: PMC6468103 DOI: 10.1523/jneurosci.2565-18.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/14/2018] [Accepted: 01/03/2019] [Indexed: 02/06/2023] Open
Abstract
GluA2-lacking Ca2+-permeable AMPARs (CP-AMPARs) play integral roles in synaptic plasticity and can mediate excitotoxic cellular signaling at glutamatergic synapses. However, the developmental profile of functional CP-AMPARs at the auditory brainstem remains poorly understood. Through a combination of electrophysiological and live-cell Ca2+ imaging from mice of either sex, we show that the synaptic release of glutamate from the calyx of Held nerve terminal activates CP-AMPARs in the principal cells of the medial nucleus of the trapezoid body in the brainstem. This leads to significant Ca2+ influx through these receptors before the onset of hearing at postnatal day 12 (P12). Using a selective open channel blocker of CP-AMPARs, IEM-1460, we estimate that ∼80% of the AMPAR population are permeable to Ca2+ at immature P4-P5 synapses. However, after the onset of hearing, Ca2+ influx through these receptors was greatly reduced. We estimate that CP-AMPARs comprise approximately 40% and 33% of the AMPAR population at P18-P22 and P30-P34, respectively. By quantifying the rate of EPSC block by IEM-1460, we found an increased heterogeneity in glutamate release probability for adult-like calyces (P30-P34). Using tetraethylammonium (TEA), a presynaptic potassium channel blocker, we show that the apparent reduction of CP-AMPARs in more mature synapses is not a consequence of presynaptic action potential (AP) speeding. Finally, through postsynaptic AP recordings, we show that inhibition of CP-AMPARs reduces spike fidelity in juvenile synapses, but not in more mature synapses. We conclude that the expression of functional CP-AMPARs declines over early postnatal development in the calyx of Held synapse.SIGNIFICANCE STATEMENT The calyx of Held synapse is pivotal to the circuitry that computes sound localization. Postsynaptic Ca2+ influx via AMPARs may be critical for signaling the maturation of this brainstem synapse. The GluA4 subunit may dominate the AMPAR complex at mature synapses because of its fast gating kinetics and large unitary conductance. The expectation is that AMPARs dominated by GluA4 subunits should be highly Ca2+ permeable. However, we find that Ca2+-permeable AMPAR expression declines during postnatal development. Using the rate of EPSC block by IEM-1460, an open channel blocker of Ca2+-permeable AMPARs, we propose a novel method to determine glutamate release probability and uncover an increased heterogeneity in release probability for more mature calyces of Held nerve terminals.
Collapse
Affiliation(s)
- Brendan Lujan
- Vollum Institute, Oregon Health and Science University, Portland, Oregon 97239
| | - Andre Dagostin
- Vollum Institute, Oregon Health and Science University, Portland, Oregon 97239
| | | |
Collapse
|
24
|
Timing constraints of action potential evoked Ca 2+ current and transmitter release at a central nerve terminal. Sci Rep 2019; 9:4448. [PMID: 30872753 PMCID: PMC6418091 DOI: 10.1038/s41598-019-41120-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 02/28/2019] [Indexed: 02/07/2023] Open
Abstract
The waveform of presynaptic action potentials (APs) regulates the magnitude of Ca2+ currents (ICa) and neurotransmitter release. However, how APs control the timing of synaptic transmission remains unclear. Using the calyx of Held synapse, we find that Na+ and K+ channels affect the timing by changing the AP waveform. Specifically, the onset of ICa depends on the repolarization but not depolarization rate of APs, being near the end of repolarization phase for narrow APs and advancing to the early repolarization phase for wide APs. Increasing AP amplitude has little effect on the activation but delays the peak time of ICa. Raising extracellular Ca2+ concentration increases the amplitude of ICa yet does not alter their onset timing. Developmental shortening of APs ensures ICa as a tail current and faithful synaptic delay, which is particularly important at the physiological temperature (35 °C) as ICa evoked by broad pseudo-APs can occur in the depolarization phase. The early onset of ICa is more prominent at 35 °C than at 22 °C, likely resulting from a temperature-dependent shift in the activation threshold and accelerated gating kinetics of Ca2+ channels. These results suggest that the timing of Ca2+ influx depends on the AP waveform dictated by voltage-gated channels and temperature.
Collapse
|
25
|
Fekete A, Nakamura Y, Yang YM, Herlitze S, Mark MD, DiGregorio DA, Wang LY. Underpinning heterogeneity in synaptic transmission by presynaptic ensembles of distinct morphological modules. Nat Commun 2019; 10:826. [PMID: 30778063 PMCID: PMC6379440 DOI: 10.1038/s41467-019-08452-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 12/28/2018] [Indexed: 11/09/2022] Open
Abstract
Synaptic heterogeneity is widely observed but its underpinnings remain elusive. We addressed this issue using mature calyx of Held synapses whose numbers of bouton-like swellings on stalks of the nerve terminals inversely correlate with release probability (Pr). We examined presynaptic Ca2+ currents and transients, topology of fluorescently tagged knock-in Ca2+ channels, and Ca2+ channel-synaptic vesicle (SV) coupling distance using Ca2+ chelator and inhibitor of septin cytomatrix in morphologically diverse synapses. We found that larger clusters of Ca2+ channels with tighter coupling distance to SVs elevate Pr in stalks, while smaller clusters with looser coupling distance lower Pr in swellings. Septin is a molecular determinant of the differences in coupling distance. Supported by numerical simulations, we propose that varying the ensemble of two morphological modules containing distinct Ca2+ channel-SV topographies diversifies Pr in the terminal, thereby establishing a morpho-functional continuum that expands the coding capacity within a single synapse population.
Collapse
Affiliation(s)
- Adam Fekete
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, 555 University Ave, Toronto, ON, M5G 1X8, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Yukihiro Nakamura
- Department of Pharmacology, Jikei University School of Medicine, Nishishinbashi, Minato-ku, Tokyo, 1058461, Japan
| | - Yi-Mei Yang
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, 555 University Ave, Toronto, ON, M5G 1X8, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Department of Biomedical Sciences, University of Minnesota Medical School, 1035 University Drive, Duluth, MN, 55812, USA
| | - Stefan Herlitze
- Department of General Zoology and Neurobiology, Ruhr-University Bochum, Universitätsstrasse 150, D-44780, Bochum, Germany
| | - Melanie D Mark
- Department of General Zoology and Neurobiology, Ruhr-University Bochum, Universitätsstrasse 150, D-44780, Bochum, Germany
| | - David A DiGregorio
- Unit of Dynamic Neuronal Imaging, Institut Pasteur, 25 rue du Dr Roux, 75724, Paris Cedex 15, France
- Centre National de la Recherche Scientifique (CNRS), UMR 3571, Genes, Synapses and Cognition, Institut Pasteur, 25 rue du Dr Roux, 75724, Paris Cedex 15, France
| | - Lu-Yang Wang
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, 555 University Ave, Toronto, ON, M5G 1X8, Canada.
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
26
|
Joris PX, Trussell LO. The Calyx of Held: A Hypothesis on the Need for Reliable Timing in an Intensity-Difference Encoder. Neuron 2018; 100:534-549. [PMID: 30408442 PMCID: PMC6263157 DOI: 10.1016/j.neuron.2018.10.026] [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: 02/02/2018] [Revised: 08/16/2018] [Accepted: 10/15/2018] [Indexed: 12/18/2022]
Abstract
The calyx of Held is the preeminent model for the study of synaptic function in the mammalian CNS. Despite much work on the synapse and associated circuit, its role in hearing remains enigmatic. We propose that the calyx is one of the key adaptations that enables an animal to lateralize transient sounds. The calyx is part of a binaural circuit that is biased toward high sound frequencies and is sensitive to intensity differences between the ears. This circuit also shows marked sensitivity to interaural time differences, but only for brief sound transients ("clicks"). In a natural environment, such transients are rare except as adventitious sounds generated by other animals moving at close range. We argue that the calyx, and associated temporal specializations, evolved to enable spatial localization of sound transients, through a neural code congruent with the circuit's sensitivity to interaural intensity differences, thereby conferring a key benefit to survival.
Collapse
Affiliation(s)
- Philip X Joris
- Laboratory of Auditory Neurophysiology, Department of Neurosciences, University of Leuven, Leuven B-3000, Belgium.
| | - Laurence O Trussell
- Oregon Hearing Research Center and Vollum Institute, Oregon Health and Science University, Portland, OR 97239, USA
| |
Collapse
|
27
|
Pre- and postsynaptic ionotropic glutamate receptors in the auditory system of mammals. Hear Res 2018; 362:1-13. [DOI: 10.1016/j.heares.2018.02.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 02/16/2018] [Accepted: 02/21/2018] [Indexed: 01/22/2023]
|
28
|
Zhang B, Seigneur E, Wei P, Gokce O, Morgan J, Südhof TC. Developmental plasticity shapes synaptic phenotypes of autism-associated neuroligin-3 mutations in the calyx of Held. Mol Psychiatry 2017; 22:1483-1491. [PMID: 27725662 PMCID: PMC5687809 DOI: 10.1038/mp.2016.157] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 07/08/2016] [Accepted: 07/27/2016] [Indexed: 01/21/2023]
Abstract
Neuroligins are postsynaptic cell-adhesion molecules that bind to presynaptic neurexins. Mutations in neuroligin-3 predispose to autism, but how such mutations affect synaptic function remains incompletely understood. Here we systematically examined the effect of three autism-associated mutations, the neuroligin-3 knockout, the R451C knockin, and the R704C knockin, on synaptic transmission in the calyx of Held, a central synapse ideally suited for high-resolution analyses of synaptic transmission. Surprisingly, germline knockout of neuroligin-3 did not alter synaptic transmission, whereas the neuroligin-3 R451C and R704C knockins decreased and increased, respectively, synaptic transmission. These puzzling results prompted us to ask whether neuroligin-3 mutant phenotypes may be reshaped by developmental plasticity. Indeed, conditional knockout of neuroligin-3 during late development produced a marked synaptic phenotype, whereas conditional knockout of neuroligin-3 during early development caused no detectable effect, mimicking the germline knockout. In canvassing potentially redundant candidate genes, we identified developmentally early expression of another synaptic neurexin ligand, cerebellin-1. Strikingly, developmentally early conditional knockout of cerebellin-1 only modestly impaired synaptic transmission, whereas in contrast to the individual single knockouts, developmentally early conditional double knockout of both cerebellin-1 and neuroligin-3 severely decreased synaptic transmission. Our data suggest an unanticipated mechanism of developmental compensation whereby cerebellin-1 and neuroligin-3 functionally occlude each other during development of calyx synapses. Thus, although acute manipulations more likely reveal basic gene functions, developmental plasticity can be a major factor in shaping the overall phenotypes of genetic neuropsychiatric disorders.
Collapse
Affiliation(s)
- B Zhang
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - E Seigneur
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - P Wei
- Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - O Gokce
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - J Morgan
- Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - TC Südhof
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
29
|
Ca 2+-Permeable AMPARs Mediate Glutamatergic Transmission and Excitotoxic Damage at the Hair Cell Ribbon Synapse. J Neurosci 2017; 37:6162-6175. [PMID: 28539424 DOI: 10.1523/jneurosci.3644-16.2017] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 04/20/2017] [Accepted: 04/24/2017] [Indexed: 01/21/2023] Open
Abstract
We report functional and structural evidence for GluA2-lacking Ca2+-permeable AMPARs (CP-AMPARs) at the mature hair cell ribbon synapse. By using the methodological advantages of three species (of either sex), we demonstrate that CP-AMPARs are present at the hair cell synapse in an evolutionarily conserved manner. Via a combination of in vivo electrophysiological and Ca2+ imaging approaches in the larval zebrafish, we show that hair cell stimulation leads to robust Ca2+ influx into afferent terminals. Prolonged application of AMPA caused loss of afferent terminal responsiveness, whereas blocking CP-AMPARs protects terminals from excitotoxic swelling. Immunohistochemical analysis of AMPAR subunits in mature rat cochlea show regions within synapses lacking the GluA2 subunit. Paired recordings from adult bullfrog auditory synapses demonstrate that CP-AMPARs mediate a major component of glutamatergic transmission. Together, our results support the importance of CP-AMPARs in mediating transmission at the hair cell ribbon synapse. Further, excess Ca2+ entry via CP-AMPARs may underlie afferent terminal damage following excitotoxic challenge, suggesting that limiting Ca2+ levels in the afferent terminal may protect against cochlear synaptopathy associated with hearing loss.SIGNIFICANCE STATEMENT A single incidence of noise overexposure causes damage at the hair cell synapse that later leads to neurodegeneration and exacerbates age-related hearing loss. A first step toward understanding cochlear neurodegeneration is to identify the cause of initial excitotoxic damage to the postsynaptic neuron. Using a combination of immunohistochemical, electrophysiological, and Ca2+ imaging approaches in evolutionarily divergent species, we demonstrate that Ca2+-permeable AMPARs (CP-AMPARs) mediate glutamatergic transmission at the adult auditory hair cell synapse. Overexcitation of the terminal causes Ca2+ accumulation and swelling that can be prevented by blocking CP-AMPARs. We demonstrate that CP-AMPARs mediate transmission at this first-order sensory synapse and that limiting Ca2+ accumulation in the terminal may protect against hearing loss.
Collapse
|
30
|
The number and distribution of AMPA receptor channels containing fast kinetic GluA3 and GluA4 subunits at auditory nerve synapses depend on the target cells. Brain Struct Funct 2017; 222:3375-3393. [PMID: 28397107 PMCID: PMC5676837 DOI: 10.1007/s00429-017-1408-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 03/20/2017] [Indexed: 02/07/2023]
Abstract
The neurotransmitter receptor subtype, number, density, and distribution relative to the location of transmitter release sites are key determinants of signal transmission. AMPA-type ionotropic glutamate receptors (AMPARs) containing GluA3 and GluA4 subunits are prominently expressed in subsets of neurons capable of firing action potentials at high frequencies, such as auditory relay neurons. The auditory nerve (AN) forms glutamatergic synapses on two types of relay neurons, bushy cells (BCs) and fusiform cells (FCs) of the cochlear nucleus. AN-BC and AN-FC synapses have distinct kinetics; thus, we investigated whether the number, density, and localization of GluA3 and GluA4 subunits in these synapses are differentially organized using quantitative freeze-fracture replica immunogold labeling. We identify a positive correlation between the number of AMPARs and the size of AN-BC and AN-FC synapses. Both types of AN synapses have similar numbers of AMPARs; however, the AN-BC have a higher density of AMPARs than AN-FC synapses, because the AN-BC synapses are smaller. A higher number and density of GluA3 subunits are observed at AN-BC synapses, whereas a higher number and density of GluA4 subunits are observed at AN-FC synapses. The intrasynaptic distribution of immunogold labeling revealed that AMPAR subunits, particularly GluA3, are concentrated at the center of the AN-BC synapses. The central distribution of AMPARs is absent in GluA3-knockout mice, and gold particles are evenly distributed along the postsynaptic density. GluA4 gold labeling was homogenously distributed along both synapse types. Thus, GluA3 and GluA4 subunits are distributed at AN synapses in a target-cell-dependent manner.
Collapse
|
31
|
García-Hernández S, Abe M, Sakimura K, Rubio ME. Impaired auditory processing and altered structure of the endbulb of Held synapse in mice lacking the GluA3 subunit of AMPA receptors. Hear Res 2016; 344:284-294. [PMID: 28011083 DOI: 10.1016/j.heares.2016.12.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 10/28/2016] [Accepted: 12/12/2016] [Indexed: 02/07/2023]
Abstract
AMPA glutamate receptor complexes with fast kinetics conferred by subunits like GluA3 and GluA4 are essential for temporal precision of synaptic transmission. The specific role of GluA3 in auditory processing and experience related changes in the auditory brainstem remain unknown. We investigated the role of the GluA3 in auditory processing by using wild type (WT) and GluA3 knockout (GluA3-KO) mice. We recorded auditory brainstem responses (ABR) to assess auditory function and used electron microscopy to evaluate the ultrastructure of the auditory nerve synapse on bushy cells (AN-BC synapse). Since labeling for GluA3 subunit increases on auditory nerve synapses within the cochlear nucleus in response to transient sound reduction, we investigated the role of GluA3 in experience-dependent changes in auditory processing. We induced transient sound reduction by plugging one ear and evaluated ABR threshold and peak amplitude recovery for up to 60 days after ear plug removal in WT and GluA3-KO mice. We found that the deletion of GluA3 leads to impaired auditory signaling that is reflected in decreased ABR peak amplitudes, an increased latency of peak 2, early onset hearing loss and reduced numbers and sizes of postsynaptic densities (PSDs) of AN-BC synapses. Additionally, the lack of GluA3 hampers ABR threshold recovery after transient ear plugging. We conclude that GluA3 is required for normal auditory signaling, normal ultrastructure of AN-BC synapses in the cochlear nucleus and normal experience-dependent changes in auditory processing after transient sound reduction.
Collapse
Affiliation(s)
- Sofía García-Hernández
- Department of Otolaryngology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Manabu Abe
- Niigata University Brain Research Institute, Japan
| | | | - María E Rubio
- Department of Otolaryngology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA; Department of Neurobiology, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA; Center for the Neural Basis of Cognition, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
32
|
Pilati N, Linley DM, Selvaskandan H, Uchitel O, Hennig MH, Kopp-Scheinpflug C, Forsythe ID. Acoustic trauma slows AMPA receptor-mediated EPSCs in the auditory brainstem, reducing GluA4 subunit expression as a mechanism to rescue binaural function. J Physiol 2016; 594:3683-703. [PMID: 27104476 PMCID: PMC4929335 DOI: 10.1113/jp271929] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 04/13/2016] [Indexed: 12/19/2022] Open
Abstract
KEY POINTS Lateral superior olive (LSO) principal neurons receive AMPA receptor (AMPAR) - and NMDA receptor (NMDAR)-mediated EPSCs and glycinergic IPSCs. Both EPSCs and IPSCs have slow kinetics in prehearing animals, which during developmental maturation accelerate to sub-millisecond decay time-constants. This correlates with a change in glutamate and glycine receptor subunit composition quantified via mRNA levels. The NMDAR-EPSCs accelerate over development to achieve decay time-constants of 2.5 ms. This is the fastest NMDAR-mediated EPSC reported. Acoustic trauma (AT, loud sounds) slow AMPAR-EPSC decay times, increasing GluA1 and decreasing GluA4 mRNA. Modelling of interaural intensity difference suggests that the increased EPSC duration after AT shifts interaural level difference to the right and compensates for hearing loss. Two months after AT the EPSC decay times recovered to control values. Synaptic transmission in the LSO matures by postnatal day 20, with EPSCs and IPSCs having fast kinetics. AT changes the AMPAR subunits expressed and slows the EPSC time-course at synapses in the central auditory system. ABSTRACT Damaging levels of sound (acoustic trauma, AT) diminish peripheral synapses, but what is the impact on the central auditory pathway? Developmental maturation of synaptic function and hearing were characterized in the mouse lateral superior olive (LSO) from postnatal day 7 (P7) to P96 using voltage-clamp and auditory brainstem responses. IPSCs and EPSCs show rapid acceleration during development, so that decay kinetics converge to similar sub-millisecond time-constants (τ, 0.87 ± 0.11 and 0.77 ± 0.08 ms, respectively) in adult mice. This correlated with LSO mRNA levels for glycinergic and glutamatergic ionotropic receptor subunits, confirming a switch from Glyα2 to Glyα1 for IPSCs and increased expression of GluA3 and GluA4 subunits for EPSCs. The NMDA receptor (NMDAR)-EPSC decay τ accelerated from >40 ms in prehearing animals to 2.6 ± 0.4 ms in adults, as GluN2C expression increased. In vivo induction of AT at around P20 disrupted IPSC and EPSC integration in the LSO, so that 1 week later the AMPA receptor (AMPAR)-EPSC decay was slowed and mRNA for GluA1 increased while GluA4 decreased. In contrast, GlyR IPSC and NMDAR-EPSC decay times were unchanged. Computational modelling confirmed that matched IPSC and EPSC kinetics are required to generate mature interaural level difference functions, and that longer-lasting EPSCs compensate to maintain binaural function with raised auditory thresholds after AT. We conclude that LSO excitatory and inhibitory synaptic drive matures to identical time-courses, that AT changes synaptic AMPARs by expression of subunits with slow kinetics (which recover over 2 months) and that loud sounds reversibly modify excitatory synapses in the brain, changing synaptic function for several weeks after exposure.
Collapse
Affiliation(s)
- Nadia Pilati
- Autifony Srl Laboratories, Medicines Research Centre, 37135, Verona, Italy.,MRC Toxicology Unit, Hodgkin Bldg, University of Leicester, Leicester, LE1 9HN, UK
| | - Deborah M Linley
- Department of Neuroscience, Psychology & Behaviour, University of Leicester, Leicester, LE1 9HN, UK
| | - Haresh Selvaskandan
- Department of Neuroscience, Psychology & Behaviour, University of Leicester, Leicester, LE1 9HN, UK
| | - Osvaldo Uchitel
- Instituto de Fisiología y Biología Molecular y Neurociencias, Universidad de Buenos Aires-CONICET, Facultad de Ciencias Exactas y Naturales, Ciudad Universitaria, C1428-Buenos Aires, Argentina
| | - Matthias H Hennig
- Institute for Adaptive and Neural Computation, School of Informatics, University of Edinburgh, Edinburgh, EH8 9AB, UK.,SynthSys, C. H. Waddington Building, The Kings Buildings Campus, Edinburgh, UK
| | - Cornelia Kopp-Scheinpflug
- MRC Toxicology Unit, Hodgkin Bldg, University of Leicester, Leicester, LE1 9HN, UK.,Department of Biology II, Ludwig-Maximilians-Universität München, Planegg-Martinsried, D-82152, Munich, Germany
| | - Ian D Forsythe
- MRC Toxicology Unit, Hodgkin Bldg, University of Leicester, Leicester, LE1 9HN, UK.,Department of Neuroscience, Psychology & Behaviour, University of Leicester, Leicester, LE1 9HN, UK
| |
Collapse
|
33
|
Blosa M, Sonntag M, Jäger C, Weigel S, Seeger J, Frischknecht R, Seidenbecher CI, Matthews RT, Arendt T, Rübsamen R, Morawski M. The extracellular matrix molecule brevican is an integral component of the machinery mediating fast synaptic transmission at the calyx of Held. J Physiol 2015. [PMID: 26223835 DOI: 10.1113/jp270849] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS The proteoglycan brevican is a major component of the extracellular matrix of perineuronal nets and is highly enriched in the perisynaptic space suggesting a role for synaptic transmission. We have introduced the calyx of Held in the auditory brainstem as a model system to study the impact of brevican on dynamics and reliability of synaptic transmission. In vivo extracellular single-unit recordings at the calyx of Held in brevican-deficient mice yielded a significant increase in the action potential (AP) transmission delay and a prolongation of pre- and postsynaptic APs. The changes in dynamics of signal transmission were accompanied by the reduction of presynaptic vGlut1 and ultrastructural changes in the perisynaptic space. These data show that brevican is an important mediator of fast synaptic transmission at the calyx of Held. ABSTRACT The extracellular matrix is an integral part of the neural tissue. Its most conspicuous manifestation in the brain are the perineuronal nets (PNs) which surround somata and proximal dendrites of distinct neuron types. The chondroitin sulfate proteoglycan brevican is a major component of PNs. In contrast to other PN-comprising proteoglycans (e.g. aggrecan and neurocan), brevican is mainly expressed in the perisynaptic space closely associated with both the pre- and postsynaptic membrane. This specific localization prompted the hypothesis that brevican might play a role in synaptic transmission. In the present study we specifically investigated the role of brevican in synaptic transmission at a central synapse, the calyx of Held in the medial nucleus of the trapezoid body, by the use of in vivo electrophysiology, immunohistochemistry, biochemistry and electron microscopy. In vivo extracellular single-unit recordings were acquired in brevican-deficient mice and the dynamics and reliability of synaptic transmission were compared to wild-type littermates. In knockout mice, the speed of pre-to-postsynaptic action potential (AP) transmission was reduced and the duration of the respective pre- and postsynaptic APs increased. The reliability of signal transmission, however, was not affected by the lack of brevican. The changes in dynamics of signal transmission were accompanied by the reduction of (i) presynaptic vGlut1 and (ii) the size of subsynaptic cavities. The present results suggest an essential role of brevican for the functionality of high-speed synaptic transmission at the calyx of Held.
Collapse
Affiliation(s)
- Maren Blosa
- Paul Flechsig Institute for Brain Research, Faculty of Medicine, University of Leipzig, 04103, Leipzig, Germany
| | - Mandy Sonntag
- Paul Flechsig Institute for Brain Research, Faculty of Medicine, University of Leipzig, 04103, Leipzig, Germany.,Institute of Biology, Faculty of Biology, Pharmacy and Psychology, University of Leipzig, 04103, Leipzig, Germany
| | - Carsten Jäger
- Paul Flechsig Institute for Brain Research, Faculty of Medicine, University of Leipzig, 04103, Leipzig, Germany
| | - Solveig Weigel
- Paul Flechsig Institute for Brain Research, Faculty of Medicine, University of Leipzig, 04103, Leipzig, Germany
| | - Johannes Seeger
- Institute of Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, University of Leipzig, 04103, Leipzig, Germany
| | | | | | - Russell T Matthews
- Department of Neuroscience and Physiology, State University of New York Upstate Medical University, Syracuse, NY, 13210, USA
| | - Thomas Arendt
- Paul Flechsig Institute for Brain Research, Faculty of Medicine, University of Leipzig, 04103, Leipzig, Germany
| | - Rudolf Rübsamen
- Institute of Biology, Faculty of Biology, Pharmacy and Psychology, University of Leipzig, 04103, Leipzig, Germany
| | - Markus Morawski
- Paul Flechsig Institute for Brain Research, Faculty of Medicine, University of Leipzig, 04103, Leipzig, Germany
| |
Collapse
|
34
|
Wright S, Hwang Y, Oertel D. Synaptic transmission between end bulbs of Held and bushy cells in the cochlear nucleus of mice with a mutation in Otoferlin. J Neurophysiol 2014; 112:3173-88. [PMID: 25253474 DOI: 10.1152/jn.00522.2014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Mice that carry a mutation in a calcium binding domain of Otoferlin, the putative calcium sensor at hair cell synapses, have normal distortion product otoacoustic emissions (DPOAEs), but auditory brain stem responses (ABRs) are absent. In mutant mice mechanotransduction is normal but transmission of acoustic information to the auditory pathway is blocked even before the onset of hearing. The cross-sectional area of the auditory nerve of mutant mice is reduced by 54%, and the volume of ventral cochlear nuclei is reduced by 46% relative to hearing control mice. While the tonotopic organization was not detectably changed in mutant mice, the axons to end bulbs of Held and the end bulbs themselves were smaller. In mutant mice bushy cells in the anteroventral cochlear nucleus (aVCN) have the electrophysiological hallmarks of control cells. Spontaneous miniature excitatory postsynaptic currents (EPSCs) occur with similar frequencies and have similar shapes in deaf as in hearing animals, but they are 24% larger in deaf mice. Bushy cells in deaf mutant mice are contacted by ∼2.6 auditory nerve fibers compared with ∼2.0 in hearing control mice. Furthermore, each fiber delivers more synaptic current, on average 4.8 nA compared with 3.4 nA, in deaf versus hearing control mice. The quantal content of evoked EPSCs is not different between mutant and control mice; the increase in synaptic current delivered in mutant mice is accounted for by the increased response to the size of the quanta. Although responses to shocks presented at long intervals are larger in mutant mice, they depress more rapidly than in hearing control mice.
Collapse
Affiliation(s)
- Samantha Wright
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin; and
| | - Youngdeok Hwang
- I.B.M. Thomas J. Watson Research Center, Yorktown Heights, New York
| | - Donata Oertel
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin; and
| |
Collapse
|
35
|
Yang YM, Wang W, Fedchyshyn MJ, Zhou Z, Ding J, Wang LY. Enhancing the fidelity of neurotransmission by activity-dependent facilitation of presynaptic potassium currents. Nat Commun 2014; 5:4564. [PMID: 25078759 PMCID: PMC4503407 DOI: 10.1038/ncomms5564] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Accepted: 07/01/2014] [Indexed: 12/02/2022] Open
Abstract
Neurons convey information in bursts of spikes across chemical synapses where the fidelity of information transfer critically depends on synaptic input-output relationship. With a limited number of synaptic vesicles (SVs) in the readily-releasable pool (RRP), how nerve terminals sustain transmitter release during intense activity remains poorly understood. Here we report that presynaptic K+ currents evoked by spikes facilitate in a Ca2+-independent but frequency- and voltage-dependent manner. Experimental evidence and computer simulations demonstrate this facilitation originates from dynamic transition of intermediate gating states of voltage-gated K+ channels (Kvs), and specifically attenuates spike amplitude and inter-spike potential during high-frequency firing. Single or paired recordings from a mammalian central synapse further reveal that facilitation of Kvs constrains presynaptic Ca2+ influx, thereby efficiently allocating SVs in the RRP to drive postsynaptic spiking at high rates. We conclude that presynaptic Kv facilitation imparts neurons with a powerful control of transmitter release to dynamically support high-fidelity neurotransmission.
Collapse
Affiliation(s)
- Yi-Mei Yang
- 1] Program in Neurosciences and Mental Health, SickKids Research Institute, Toronto, Ontario, Canada M5G 1X8 [2] Department of Physiology, University of Toronto, Toronto, Ontario, Canada M5S 1A8 [3]
| | - Wei Wang
- 1] Key Laboratory of Molecular Biophysics of the Ministry of Education, Huazhong University of Science and Technology, Wuhan 430074, China [2] Department of Medical Engineering, the 180th Hospital of PLA, Quanzhou 362000, China [3]
| | - Michael J Fedchyshyn
- 1] Program in Neurosciences and Mental Health, SickKids Research Institute, Toronto, Ontario, Canada M5G 1X8 [2] Department of Physiology, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| | - Zhuan Zhou
- Institute of Molecular Medicine &PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Jiuping Ding
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lu-Yang Wang
- 1] Program in Neurosciences and Mental Health, SickKids Research Institute, Toronto, Ontario, Canada M5G 1X8 [2] Department of Physiology, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| |
Collapse
|
36
|
Roberts MT, Seeman SC, Golding NL. The relative contributions of MNTB and LNTB neurons to inhibition in the medial superior olive assessed through single and paired recordings. Front Neural Circuits 2014; 8:49. [PMID: 24860434 PMCID: PMC4030206 DOI: 10.3389/fncir.2014.00049] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 04/24/2014] [Indexed: 02/02/2023] Open
Abstract
The medial superior olive (MSO) senses microsecond differences in the coincidence of binaural signals, a critical cue for detecting sound location along the azimuth. An important component of this circuit is provided by inhibitory neurons of the medial and lateral nuclei of the trapezoid body (MNTB and LNTB, respectively). While MNTB neurons are fairly well described, little is known about the physiology of LNTB neurons. Using whole cell recordings from gerbil brainstem slices, we found that LNTB and MNTB neurons have similar membrane time constants and input resistances and fire brief action potentials, but only LNTB neurons fire repetitively in response to current steps. We observed that LNTB neurons receive graded excitatory and inhibitory synaptic inputs, with at least some of the latter arriving from other LNTB neurons. To address the relative timing of inhibition to the MSO from the LNTB versus the MNTB, we examined inhibitory responses to auditory nerve stimulation using a slice preparation that retains the circuitry from the auditory nerve to the MSO intact. Despite the longer physical path length of excitatory inputs driving contralateral inhibition, inhibition from both pathways arrived with similar latency and jitter. An analysis of paired whole cell recordings between MSO and MNTB neurons revealed a short and reliable delay between the action potential peak in MNTB neurons and the onset of the resulting IPSP (0.55 ± 0.01 ms, n = 4, mean ± SEM). Reconstructions of biocytin-labeled neurons showed that MNTB axons ranged from 580 to 858 μm in length (n = 4). We conclude that while both LNTB and MNTB neurons provide similarly timed inhibition to MSO neurons, the reliability of inhibition from the LNTB at higher frequencies is more constrained relative to that from the MNTB due to differences in intrinsic properties, the strength of excitatory inputs, and the presence of feedforward inhibition.
Collapse
Affiliation(s)
- Michael T Roberts
- Department of Neuroscience, Center for Learning and Memory, The University of Texas at Austin Austin, TX, USA
| | - Stephanie C Seeman
- Department of Neuroscience, Center for Learning and Memory, The University of Texas at Austin Austin, TX, USA
| | - Nace L Golding
- Department of Neuroscience, Center for Learning and Memory, The University of Texas at Austin Austin, TX, USA
| |
Collapse
|
37
|
Morphological and physiological development of auditory synapses. Hear Res 2014; 311:3-16. [PMID: 24508369 DOI: 10.1016/j.heares.2014.01.007] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Revised: 01/13/2014] [Accepted: 01/20/2014] [Indexed: 02/07/2023]
Abstract
Acoustic communication requires gathering, transforming, and interpreting diverse sound cues. To achieve this, all the spatial and temporal features of complex sound stimuli must be captured in the firing patterns of the primary sensory neurons and then accurately transmitted along auditory pathways for additional processing. The mammalian auditory system relies on several synapses with unique properties in order to meet this task: the auditory ribbon synapses, the endbulb of Held, and the calyx of Held. Each of these synapses develops morphological and electrophysiological characteristics that enable the remarkably precise signal transmission necessary for conveying the miniscule differences in timing that underly sound localization. In this article, we review the current knowledge of how these synapses develop and mature to acquire the specialized features necessary for the sense of hearing.
Collapse
|
38
|
Xiao L, Michalski N, Kronander E, Gjoni E, Genoud C, Knott G, Schneggenburger R. BMP signaling specifies the development of a large and fast CNS synapse. Nat Neurosci 2013; 16:856-64. [PMID: 23708139 DOI: 10.1038/nn.3414] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Accepted: 05/02/2013] [Indexed: 11/09/2022]
Abstract
Large excitatory synapses with multiple active zones ensure reliable and fast information transfer at specific points in neuronal circuits. However, the mechanisms that determine synapse size in CNS circuits are largely unknown. Here we use the calyx of Held synapse, a major relay in the auditory system, to identify and study signaling pathways that specify large nerve terminal size and fast synaptic transmission. Using genome-wide screening, we identified bone morphogenetic proteins (BMPs) as candidate signaling molecules in the area of calyx synapses. Conditional deletion of BMP receptors in the auditory system of mice led to aberrations of synapse morphology and function specifically at the calyx of Held, with impaired nerve terminal growth, loss of monoinnervation and less mature transmitter release properties. Thus, BMP signaling specifies large and fast-transmitting synapses in the auditory system in a process that shares homologies with, but also extends beyond, retrograde BMP signaling at Drosophila neuromuscular synapses.
Collapse
Affiliation(s)
- Le Xiao
- Laboratory of Synaptic Mechanisms, Brain Mind Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | | | | | | | | | | | | |
Collapse
|
39
|
Lina IA, Lauer AM. Rapid measurement of auditory filter shape in mice using the auditory brainstem response and notched noise. Hear Res 2013; 298:73-9. [PMID: 23347916 PMCID: PMC3639490 DOI: 10.1016/j.heares.2013.01.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Revised: 12/27/2012] [Accepted: 01/07/2013] [Indexed: 11/21/2022]
Abstract
The notched noise method is an effective procedure for measuring frequency resolution and auditory filter shapes in both human and animal models of hearing. Briefly, auditory filter shape and bandwidth estimates are derived from masked thresholds for tones presented in noise containing widening spectral notches. As the spectral notch widens, increasingly less of the noise falls within the auditory filter and the tone becomes more detectible until the notch width exceeds the filter bandwidth. Behavioral procedures have been used for the derivation of notched noise auditory filter shapes in mice; however, the time and effort needed to train and test animals on these tasks renders a constraint on the widespread application of this testing method. As an alternative procedure, we combined relatively non-invasive auditory brainstem response (ABR) measurements and the notched noise method to estimate auditory filters in normal-hearing mice at center frequencies of 8, 11.2, and 16 kHz. A complete set of simultaneous masked thresholds for a particular tone frequency were obtained in about an hour. ABR-derived filter bandwidths broadened with increasing frequency, consistent with previous studies. The ABR notched noise procedure provides a fast alternative to estimating frequency selectivity in mice that is well-suited to high through-put or time-sensitive screening.
Collapse
Affiliation(s)
- Ioan A. Lina
- Johns Hopkins University School of Medicine, Department of Otolaryngology – HNS, Center for Hearing and Balance, 515 Traylor, 720 Rutland Ave., Baltimore, MD 21205, United States
| | - Amanda M. Lauer
- Johns Hopkins University School of Medicine, Department of Otolaryngology – HNS, Center for Hearing and Balance, 515 Traylor, 720 Rutland Ave., Baltimore, MD 21205, United States
| |
Collapse
|
40
|
Jurgens CWD, Bell KA, McQuiston AR, Guido W. Optogenetic stimulation of the corticothalamic pathway affects relay cells and GABAergic neurons differently in the mouse visual thalamus. PLoS One 2012; 7:e45717. [PMID: 23029198 PMCID: PMC3447820 DOI: 10.1371/journal.pone.0045717] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 08/23/2012] [Indexed: 12/11/2022] Open
Abstract
The dorsal lateral geniculate nucleus (dLGN) serves as the primary conduit of retinal information to visual cortex. In addition to retinal input, dLGN receives a large feedback projection from layer VI of visual cortex. Such input modulates thalamic signal transmission in different ways that range from gain control to synchronizing network activity in a stimulus-specific manner. However, the mechanisms underlying such modulation have been difficult to study, in part because of the complex circuitry and diverse cell types this pathway innervates. To address this and overcome some of the technical limitations inherent in studying the corticothalamic (CT) pathway, we adopted a slice preparation in which we were able to stimulate CT terminal arbors in the visual thalamus of the mouse with blue light by using an adeno-associated virus to express the light-gated ion channel, ChIEF, in layer VI neurons. To examine the postsynaptic responses evoked by repetitive CT stimulation, we recorded from identified relay cells in dLGN, as well as GFP expressing GABAergic neurons in the thalamic reticular nucleus (TRN) and intrinsic interneurons of dLGN. Relay neurons exhibited large glutamatergic responses that continued to increase in amplitude with each successive stimulus pulse. While excitatory responses were apparent at postnatal day 10, the strong facilitation noted in adult was not observed until postnatal day 21. GABAergic neurons in TRN exhibited large initial excitatory responses that quickly plateaued during repetitive stimulation, indicating that the degree of facilitation was much larger for relay cells than for TRN neurons. The responses of intrinsic interneurons were smaller and took the form of a slow depolarization. These differences in the pattern of excitation for different thalamic cell types should help provide a framework for understanding how CT feedback alters the activity of visual thalamic circuitry during sensory processing as well as different behavioral or pathophysiological states.
Collapse
Affiliation(s)
- Chris W. D. Jurgens
- Department of Anatomy and Neurobiology, Virginia Commonwealth University Medical Center, Richmond, Virginia, United States of America
| | - Karen A. Bell
- Department of Anatomy and Neurobiology, Virginia Commonwealth University Medical Center, Richmond, Virginia, United States of America
| | - A. Rory McQuiston
- Department of Anatomy and Neurobiology, Virginia Commonwealth University Medical Center, Richmond, Virginia, United States of America
| | - William Guido
- Department of Anatomy and Neurobiology, Virginia Commonwealth University Medical Center, Richmond, Virginia, United States of America
- * E-mail:
| |
Collapse
|
41
|
Abstract
Some neurons in the mammalian auditory system are able to detect and report the coincident firing of inputs with remarkable temporal precision. A strong, low-voltage-activated potassium conductance (g(KL)) at the cell body and dendrites gives these neurons sensitivity to the rate of depolarization by EPSPs, allowing neurons to assess the coincidence of the rising slopes of unitary EPSPs. Two groups of neurons in the brain stem, octopus cells in the posteroventral cochlear nucleus and principal cells of the medial superior olive (MSO), extract acoustic information by assessing coincident firing of their inputs over a submillisecond timescale and convey that information at rates of up to 1000 spikes s(-1). Octopus cells detect the coincident activation of groups of auditory nerve fibres by broadband transient sounds, compensating for the travelling wave delay by dendritic filtering, while MSO neurons detect coincident activation of similarly tuned neurons from each of the two ears through separate dendritic tufts. Each makes use of filtering that is introduced by the spatial distribution of inputs on dendrites.
Collapse
Affiliation(s)
- Nace L Golding
- Section of Neurobiology and Center for Learning and Memory, University of Texas at Austin, Austin, TX, USA
| | | |
Collapse
|
42
|
Zheng Z, Sabirzhanov B, Keifer J. Two-stage AMPA receptor trafficking in classical conditioning and selective role for glutamate receptor subunit 4 (tGluA4) flop splice variant. J Neurophysiol 2012; 108:101-11. [PMID: 22490558 DOI: 10.1152/jn.01097.2011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Previously, we proposed a two-stage model for an in vitro neural correlate of eyeblink classical conditioning involving the initial synaptic incorporation of glutamate receptor A1 (GluA1)-containing α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid type receptors (AMPARs) followed by delivery of GluA4-containing AMPARs that support acquisition of conditioned responses. To test specific elements of our model for conditioning, selective knockdown of GluA4 AMPAR subunits was used using small-interfering RNAs (siRNAs). Recently, we sequenced and characterized the GluA4 subunit and its splice variants from pond turtles, Trachemys scripta elegans (tGluA4). Analysis of the relative abundance of mRNA expression by real-time RT-PCR showed that the flip/flop variants of tGluA4, tGluA4c, and a novel truncated variant tGluA4trc1 are major isoforms in the turtle brain. Here, transfection of in vitro brain stem preparations with anti-tGluA4 siRNA suppressed conditioning, tGluA4 mRNA and protein expression, and synaptic delivery of tGluA4-containing AMPARs but not tGluA1 subunits. Significantly, transfection of abducens motor neurons by nerve injections of tGluA4 flop rescue plasmid prior to anti-tGluA4 siRNA application restored conditioning and synaptic incorporation of tGluA4-containing AMPARs. In contrast, treatment with rescue plasmids for tGluA4 flip or tGluA4trc1 failed to rescue conditioning. Finally, treatment with a siRNA directed against GluA1 subunits inhibited conditioning and synaptic delivery of tGluA1-containing AMPARs and importantly, those containing tGluA4. These data strongly support our two-stage model of conditioning and our hypothesis that synaptic incorporation of tGluA4-containing AMPARs underlies the acquisition of in vitro classical conditioning. Furthermore, they suggest that tGluA4 flop may have a critical role in conditioning mechanisms compared with the other tGluA4 splice variants.
Collapse
Affiliation(s)
- Zhaoqing Zheng
- Neuroscience Group, Division of Basic Biomedical Sciences, University of South Dakota, Sanford School of Medicine, Vermillion, SD 57069, USA
| | | | | |
Collapse
|
43
|
Abstract
The calyx of Held is an axosomatic terminal in the auditory brainstem that has attracted anatomists because of its giant size and physiologists because of its accessibility to patch-clamp recordings. The calyx allows the principal neurons in the medial nucleus of the trapezoid body (MNTB) to provide inhibition that is both well timed and sustained to many other auditory nuclei. The special adaptations that allow the calyx to drive its principal neuron even when frequencies are high include a large number of release sites with low release probability, a large readily releasable pool, fast presynaptic calcium clearance and little delayed release, a large quantal size, and fast AMPA-type glutamate receptors. The transformation from a synapse that is unremarkable except for its giant size into a fast and reliable auditory relay happens in just a few days. In rodents this transformation is essentially ready when hearing starts.
Collapse
Affiliation(s)
- J Gerard G Borst
- Department of Neuroscience, Erasmus MC, University Medical Center, 3015 GE Rotterdam, The Netherlands.
| | | |
Collapse
|
44
|
Oertel D. GluA4 sustains sensing of sounds through stable, speedy, sumptuous, spineless synapses. J Physiol 2011; 589:4089-90. [PMID: 21885443 DOI: 10.1113/jphysiol.2011.214536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Donata Oertel
- Department of Neuroscience, School ofMedicine and Public Health, University of Wisconsin, Madison, Wisconsin,USA.
| |
Collapse
|