1
|
Doorn N, van Putten MJAM, Frega M. Automated inference of disease mechanisms in patient-hiPSC-derived neuronal networks. Commun Biol 2025; 8:768. [PMID: 40394269 DOI: 10.1038/s42003-025-08209-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 05/12/2025] [Indexed: 05/22/2025] Open
Abstract
Human induced pluripotent stem cells (hiPSCs)-derived neuronal networks on multi-electrode arrays (MEAs) are a powerful tool for studying neurological disorders. The electric activity patterns of these networks differ between healthy and patient-derived neurons, reflecting underlying pathology. However, elucidating these underlying molecular mechanisms requires strenuous additional experiments. Computational models can link observable network activity to underlying mechanisms by estimating biophysical model parameters that simulate the experimental observations, but this is challenging. Here, we address this challenge using simulation-based inference (SBI), a machine-learning approach, to automatically estimate all model parameters that can explain network activity. We show how SBI can accurately estimate parameters that replicate the activity of healthy hiPSC-derived neuronal networks, pinpoint molecular mechanisms affected by pharmacological agents, and identify key disease mechanisms in patient-derived neuronal networks. This demonstrates SBI's potential to automate and enhance the discovery of in vitro disease mechanisms from MEA measurements, advancing research with hiPSC-derived neuronal networks.
Collapse
Affiliation(s)
- Nina Doorn
- Department of Clinical Neurophysiology, University of Twente, Enschede, The Netherlands.
| | - Michel J A M van Putten
- Department of Clinical Neurophysiology, University of Twente, Enschede, The Netherlands
- Department of Neurology and Clinical Neurophysiology, Medisch Spectrum Twente, Enschede, The Netherlands
| | - Monica Frega
- Department of Clinical Neurophysiology, University of Twente, Enschede, The Netherlands
- Department of Informatics, Bioengineering, Robotics and Systems Engineering, University of Genoa, Genoa, Italy
| |
Collapse
|
2
|
Milanick W, Li J, Thomas CI, Al-Yaari M, Guerrero-Given D, Kamasawa N, Young SM. Presynaptic α 2δs specify synaptic gain, not synaptogenesis, in the mammalian brain. Neuron 2025:S0896-6273(25)00296-X. [PMID: 40367942 DOI: 10.1016/j.neuron.2025.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 03/13/2025] [Accepted: 04/14/2025] [Indexed: 05/16/2025]
Abstract
The α2δs are a family of extracellular synaptic molecules that are auxiliary subunits of voltage-gated Ca2+ channel (CaV) complexes. They are linked to brain disorders and are drug targets. The α2δs are implicated in controlling synapse development and function through distinct CaV-dependent and CaV-independent pathways. However, the mechanisms of action remain enigmatic since synapses contain mixtures of α2δ isoforms in the pre- and postsynaptic compartments. We developed a triple conditional knockout mouse model and demonstrated the combined selective presynaptic ablation of α2δs in vivo in a developing mammalian glutamatergic synapse. We identified presynaptic α2δs as positive regulators of Munc13-1 levels, an essential neurotransmitter release protein. We found that mammalian synapse development, presynaptic CaV2.1 organization, and the transsynaptic alignment of presynaptic release sites and postsynaptic glutamate receptors are independent of presynaptic α2δs. Therefore, our results define presynaptic α2δ regulatory roles and suggest a new α2δ role in controlling synaptic strength and plasticity.
Collapse
Affiliation(s)
- William Milanick
- Gene Therapy Center, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA; Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, IA 52242, USA
| | - Jianing Li
- Gene Therapy Center, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Connon I Thomas
- The Imaging Center and Electron Microscopy Core Facility, Max Planck Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Mohammed Al-Yaari
- Gene Therapy Center, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA
| | - Debbie Guerrero-Given
- The Imaging Center and Electron Microscopy Core Facility, Max Planck Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Naomi Kamasawa
- The Imaging Center and Electron Microscopy Core Facility, Max Planck Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Samuel M Young
- Gene Therapy Center, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pediatrics, Department of Pharmacology, University of North Carolina-Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
3
|
Hommersom MP, Doorn N, Puvogel S, Lewerissa EI, Mordelt A, Ciptasari U, Kampshoff F, Dillen L, van Beusekom E, Oudakker A, Kogo N, Dolga AM, Frega M, Schubert D, van de Warrenburg BPC, Nadif Kasri N, van Bokhoven H. CACNA1A haploinsufficiency leads to reduced synaptic function and increased intrinsic excitability. Brain 2025; 148:1286-1301. [PMID: 39460936 PMCID: PMC11969466 DOI: 10.1093/brain/awae330] [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: 02/16/2024] [Revised: 08/30/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Haploinsufficiency of CACNA1A, encoding the pore-forming α1 subunit of P/Q-type voltage-gated calcium channels, is associated with a clinically variable phenotype ranging from cerebellar ataxia to neurodevelopmental syndromes with epilepsy and intellectual disability. To understand the pathological mechanisms of CACNA1A loss-of-function variants, we characterized a human neuronal model for CACNA1A haploinsufficiency by differentiating isogenic induced pluripotent stem cell lines into glutamatergic neurons and investigated the effect of CACNA1A haploinsufficiency on mature neuronal networks through a combination of electrophysiology, gene expression analysis and in silico modelling. We observed an altered network synchronization in CACNA1A+/- networks alongside synaptic deficits, notably marked by an augmented contribution of GluA2 subunit-lacking α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. Intriguingly, these synaptic perturbations coexisted with increased non-synaptically driven activity, as characterized by inhibition of N-methyl-D-aspartate and AMPA receptors on micro-electrode arrays. Single-cell electrophysiology and gene expression analysis corroborated this increased intrinsic excitability through reduced potassium channel function and expression. Moreover, we observed partial mitigation of the CACNA1A+/- network phenotype by 4-aminopyridine, a therapeutic intervention for episodic ataxia type 2. Positive modulation of small conductance calcium-activated potassium channels could reverse the CACNA1A+/- network electrophysiological phenotype. In summary, our study pioneers the characterization of a human induced pluripotent stem cell-derived neuronal model for CACNA1A haploinsufficiency and has unveiled new mechanistic insights. Beyond showcasing synaptic deficits, this neuronal model exhibited increased intrinsic excitability mediated by diminished potassium channel function, underscoring its potential as a therapeutic discovery platform with predictive validity.
Collapse
Affiliation(s)
- Marina P Hommersom
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen 6500 HB, The Netherlands
| | - Nina Doorn
- Department of Clinical Neurophysiology, University of Twente, Enschede 7522 NB, The Netherlands
| | - Sofía Puvogel
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen 6500 HB, The Netherlands
| | - Elly I Lewerissa
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen 6500 HB, The Netherlands
| | - Annika Mordelt
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen 6500 HB, The Netherlands
| | - Ummi Ciptasari
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen 6500 HB, The Netherlands
| | - Franziska Kampshoff
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen 6500 HB, The Netherlands
| | - Lieke Dillen
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen 6500 HB, The Netherlands
| | - Ellen van Beusekom
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen 6500 HB, The Netherlands
| | - Astrid Oudakker
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen 6500 HB, The Netherlands
| | - Naoki Kogo
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen 6500 HB, The Netherlands
| | - Amalia M Dolga
- Department of Pathology and Medical Biology, Division of Pathology, University of Groningen, University Medical Center Groningen, Groningen 9713 GZ, The Netherlands
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen 9713 AV, The Netherlands
| | - Monica Frega
- Department of Clinical Neurophysiology, University of Twente, Enschede 7522 NB, The Netherlands
| | - Dirk Schubert
- Department of Cognitive Neurosciences, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen 6500 HB, The Netherlands
| | - Bart P C van de Warrenburg
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen 6500 HB, The Netherlands
| | - Nael Nadif Kasri
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen 6500 HB, The Netherlands
- Department of Cognitive Neurosciences, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen 6500 HB, The Netherlands
| | - Hans van Bokhoven
- Department of Human Genetics, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen 6500 HB, The Netherlands
- Department of Cognitive Neurosciences, Radboud University Medical Center, Donders Institute for Brain, Cognition, and Behaviour, Nijmegen 6500 HB, The Netherlands
| |
Collapse
|
4
|
Martín‐Belmonte A, Aguado C, Alfaro‐Ruiz R, Kulik A, de la Ossa L, Moreno‐Martínez AE, Alberquilla S, García‐Carracedo L, Fernández M, Fajardo‐Serrano A, Aso E, Shigemoto R, Martín ED, Fukazawa Y, Ciruela F, Luján R. Nanoarchitecture of Ca V2.1 channels and GABA B receptors in the mouse hippocampus: Impact of APP/PS1 pathology. Brain Pathol 2025; 35:e13279. [PMID: 38887180 PMCID: PMC11835447 DOI: 10.1111/bpa.13279] [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: 02/06/2024] [Accepted: 05/27/2024] [Indexed: 06/20/2024] Open
Abstract
Voltage-gated CaV2.1 (P/Q-type) Ca2+ channels play a crucial role in regulating neurotransmitter release, thus contributing to synaptic plasticity and to processes such as learning and memory. Despite their recognized importance in neural function, there is limited information on their potential involvement in neurodegenerative conditions such as Alzheimer's disease (AD). Here, we aimed to explore the impact of AD pathology on the density and nanoscale compartmentalization of CaV2.1 channels in the hippocampus in association with GABAB receptors. Histoblotting experiments showed that the density of CaV2.1 channel was significantly reduced in the hippocampus of APP/PS1 mice in a laminar-dependent manner. CaV2.1 channel was enriched in the active zone of the axon terminals and was present at a very low density over the surface of dendritic tree of the CA1 pyramidal cells, as shown by quantitative SDS-digested freeze-fracture replica labelling (SDS-FRL). In APP/PS1 mice, the density of CaV2.1 channel in the active zone was significantly reduced in the strata radiatum and lacunosum-moleculare, while it remained unaltered in the stratum oriens. The decline in Cav2.1 channel density was found to be associated with a corresponding impairment in the GABAergic synaptic function, as evidenced by electrophysiological experiments carried out in the hippocampus of APP/PS1 mice. Remarkably, double SDS-FRL showed a co-clustering of CaV2.1 channel and GABAB1 receptor in nanodomains (~40-50 nm) in wild type mice, while in APP/PS1 mice this nanoarchitecture was absent. Together, these findings suggest that the AD pathology-induced reduction in CaV2.1 channel density and CaV2.1-GABAB1 de-clustering may play a role in the synaptic transmission alterations shown in the AD hippocampus. Therefore, uncovering these layer-dependent changes in P/Q calcium currents associated with AD pathology can benefit the development of future strategies for AD management.
Collapse
Affiliation(s)
- Alejandro Martín‐Belmonte
- Departamento de Ciencias Médicas, Facultad de Medicina, Synaptic Structure Laboratory, Instituto de Biomedicina de la UCLM (IB‐UCLM)Universidad Castilla‐La ManchaAlbaceteSpain
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of NeurosciencesUniversity of BarcelonaBarcelonaSpain
- Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de BellvitgeBarcelonaSpain
| | - Carolina Aguado
- Departamento de Ciencias Médicas, Facultad de Medicina, Synaptic Structure Laboratory, Instituto de Biomedicina de la UCLM (IB‐UCLM)Universidad Castilla‐La ManchaAlbaceteSpain
- Laboratorio de Estructura Sináptica, Instituto de Investigación Sanitaria de Castilla‐La Mancha (IDISCAM)AlbaceteSpain
| | - Rocío Alfaro‐Ruiz
- Departamento de Ciencias Médicas, Facultad de Medicina, Synaptic Structure Laboratory, Instituto de Biomedicina de la UCLM (IB‐UCLM)Universidad Castilla‐La ManchaAlbaceteSpain
- Laboratorio de Estructura Sináptica, Instituto de Investigación Sanitaria de Castilla‐La Mancha (IDISCAM)AlbaceteSpain
| | - Akos Kulik
- Institute for Physiology II, Medical FacultyUniversity of FreiburgFreiburgGermany
| | - Luis de la Ossa
- Departamento de Sistemas Informáticos, Escuela Superior de Ingeniería InformáticaUniversidad de Castilla‐La ManchaAlbaceteSpain
| | - Ana Esther Moreno‐Martínez
- Departamento de Ciencias Médicas, Facultad de Medicina, Synaptic Structure Laboratory, Instituto de Biomedicina de la UCLM (IB‐UCLM)Universidad Castilla‐La ManchaAlbaceteSpain
- Laboratorio de Estructura Sináptica, Instituto de Investigación Sanitaria de Castilla‐La Mancha (IDISCAM)AlbaceteSpain
| | - Samuel Alberquilla
- Laboratory of Neurophysiology and Synaptic Plasticity, Instituto Cajal, Consejo Superior de Investigaciones CientíficasMadridSpain
| | - Lucía García‐Carracedo
- Laboratory of Neurophysiology and Synaptic Plasticity, Instituto Cajal, Consejo Superior de Investigaciones CientíficasMadridSpain
| | - Miriam Fernández
- Departamento de Ciencias Médicas, Facultad de Medicina, Synaptic Structure Laboratory, Instituto de Biomedicina de la UCLM (IB‐UCLM)Universidad Castilla‐La ManchaAlbaceteSpain
- Laboratorio de Estructura Sináptica, Instituto de Investigación Sanitaria de Castilla‐La Mancha (IDISCAM)AlbaceteSpain
| | - Ana Fajardo‐Serrano
- Departamento de Ciencias Médicas, Facultad de Medicina, Synaptic Structure Laboratory, Instituto de Biomedicina de la UCLM (IB‐UCLM)Universidad Castilla‐La ManchaAlbaceteSpain
| | - Ester Aso
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of NeurosciencesUniversity of BarcelonaBarcelonaSpain
- Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de BellvitgeBarcelonaSpain
| | - Ryuichi Shigemoto
- Institute of Science and Technology Austria (ISTA)KlosterneuburgAustria
| | - Eduardo D. Martín
- Laboratory of Neurophysiology and Synaptic Plasticity, Instituto Cajal, Consejo Superior de Investigaciones CientíficasMadridSpain
| | - Yugo Fukazawa
- Division of Brain Structure and Function, Faculty of Medical ScienceUniversity of FukuiFukuiJapan
- Life Science Innovation CenterUniversity of FukuiFukuiJapan
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of NeurosciencesUniversity of BarcelonaBarcelonaSpain
- Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de BellvitgeBarcelonaSpain
| | - Rafael Luján
- Departamento de Ciencias Médicas, Facultad de Medicina, Synaptic Structure Laboratory, Instituto de Biomedicina de la UCLM (IB‐UCLM)Universidad Castilla‐La ManchaAlbaceteSpain
- Laboratorio de Estructura Sináptica, Instituto de Investigación Sanitaria de Castilla‐La Mancha (IDISCAM)AlbaceteSpain
| |
Collapse
|
5
|
Mammadova D, Kraus C, Leis T, Popp B, Zweier C, Reis A, Trollmann R. Intrafamilial neurological phenotypic variability due to either biallelic or monoallelic pathogenic variants in CACNA1A. Front Neurol 2024; 15:1458109. [PMID: 39416668 PMCID: PMC11479977 DOI: 10.3389/fneur.2024.1458109] [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: 07/01/2024] [Accepted: 09/09/2024] [Indexed: 10/19/2024] Open
Abstract
Pathogenic heterozygous variants in CACNA1A are associated with familial hemiplegic migraine, episodic ataxia type 2 and spinocerebellar ataxia type 6, and more recently, neurodevelopmental disorders. We describe a severe, early-onset phenotype including severe muscular hypotonia, early-onset epileptic seizures, apnoea, optic atrophy and dysphagia in three siblings carrying compound heterozygous frameshift variants in CACNA1A. Two male patients died at the age of 5 or 14 months of suspected SIDS or severe developmental epileptic encephalopathy (DEE) with refractory seizures and apnoea. A male child (index patient) developed severe early-onset DEE including seizures and ictal apnoea at the age of 4 weeks. Another male child developed generalized epilepsy and mild intellectual impairment in late infancy, and his mother and his maternal uncle were identified as carriers of a known CACNA1A pathogenic variant [c.2602delG heterozygous, p. (Ala868Profs*24)] with a diagnosis of episodic ataxia type 2. This maternal pathogenic variant c.2602delG was detected in the index patient and child 2. Trio-Exome sequencing identified an additional heterozygous pathogenic variant in the CACNA1A gene, c.5476delC, p.(His1826Thrfs*30) in the index patient and child 2, which was inherited from the asymptomatic father. In conclusion, the novel compound heterozygosity for two frameshift pathogenic variants, maternally [c.2602delG, p.(Ala868Profs*24)] and paternally [c.5476delC, p.(His1826Thrfs*3)] is associated with a severe phenotype of early-onset DEE. This observation highlights the necessity of additional analyses to clarify unusual phenotypes even if a pathogenic variant has already been identified, and expands the clinical spectrum of CACNA1A-related disorders.
Collapse
Affiliation(s)
- Dilbar Mammadova
- Department of Pediatrics, Pediatric Neurology, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Cornelia Kraus
- Institute of Human Genetics, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Leis
- Department of Pediatrics, Pediatric Neurology, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Bernt Popp
- Institute of Human Genetics, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
- Center of Functional Genomics, Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Christiane Zweier
- Institute of Human Genetics, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Human Genetics, Inselspital, University of Bern, Bern, Switzerland
| | - Andre Reis
- Institute of Human Genetics, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
- Centre for Rare Disorders Erlangen, University Hospital Erlangen, Erlangen, Germany
| | - Regina Trollmann
- Department of Pediatrics, Pediatric Neurology, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
- Centre for Rare Disorders Erlangen, University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
6
|
Zhu CH, Yu JY, Ma Y, Dong Y, Wu ZY. Progressive Ataxia due to de novo Missense Variants in the CACNA1A Gene. CEREBELLUM (LONDON, ENGLAND) 2024; 23:2197-2204. [PMID: 38869769 DOI: 10.1007/s12311-024-01710-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/04/2024] [Indexed: 06/14/2024]
Abstract
The CACNA1A gene encodes the alpha-1A subunit of P/Q type voltage-gated calcium channel Cav2.1, which is associated with a broad clinical spectrum and variable symptomatology. While few patients with progressive ataxia caused by CACNA1A missense variants have been reported, here we report three unrelated Chinese patients with progressive ataxia due to de novo missense variants in the CACNA1A gene, including a novel pathogenic variant (c.4999C > G) and a previously reported pathogenic variant (c.4037G > A). Our findings and a systematic literature review show the unique phenotype of progressive ataxia caused by missense variants and enlarge the genetic and clinical spectrum of CACNA1A. This suggests that in addition to routine screening for dynamic mutations, screening for CACNA1A variants is important for clinicians facing patients with progressive ataxia.
Collapse
Affiliation(s)
- Chen-Hao Zhu
- Department of Medical Genetics and Center for Rare Diseases and Department of Neurology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Rd, Hangzhou, 310009, China
| | - Jin-Yang Yu
- Department of Medical Genetics and Center for Rare Diseases and Department of Neurology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Rd, Hangzhou, 310009, China
| | - Yin Ma
- Department of Medical Genetics and Center for Rare Diseases and Department of Neurology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Rd, Hangzhou, 310009, China
| | - Yi Dong
- Department of Medical Genetics and Center for Rare Diseases and Department of Neurology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Rd, Hangzhou, 310009, China
| | - Zhi-Ying Wu
- Department of Medical Genetics and Center for Rare Diseases and Department of Neurology, the Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Rd, Hangzhou, 310009, China.
- Nanhu Brain-Computer Interface Institute, Hangzhou, China.
- MOE Frontier Science Center for Brain Science and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
7
|
Zhou YS, Tao HB, Lv SS, Liang KQ, Shi WY, Liu KY, Li YY, Chen LY, Zhou L, Yin SJ, Zhao QR. Effects of Kv1.3 knockout on pyramidal neuron excitability and synaptic plasticity in piriform cortex of mice. Acta Pharmacol Sin 2024; 45:2045-2060. [PMID: 38862816 PMCID: PMC11420205 DOI: 10.1038/s41401-024-01275-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 03/24/2024] [Indexed: 06/13/2024]
Abstract
Kv1.3 belongs to the voltage-gated potassium (Kv) channel family, which is widely expressed in the central nervous system and associated with a variety of neuropsychiatric disorders. Kv1.3 is highly expressed in the olfactory bulb and piriform cortex and involved in the process of odor perception and nutrient metabolism in animals. Previous studies have explored the function of Kv1.3 in olfactory bulb, while the role of Kv1.3 in piriform cortex was less known. In this study, we investigated the neuronal changes of piriform cortex and feeding behavior after smell stimulation, thus revealing a link between the olfactory sensation and body weight in Kv1.3 KO mice. Coronal slices including the anterior piriform cortex were prepared, whole-cell recording and Ca2+ imaging of pyramidal neurons were conducted. We showed that the firing frequency evoked by depolarization pulses and Ca2+ influx evoked by high K+ solution were significantly increased in pyramidal neurons of Kv1.3 knockout (KO) mice compared to WT mice. Western blotting and immunofluorescence analyses revealed that the downstream signaling molecules CaMKII and PKCα were activated in piriform cortex of Kv1.3 KO mice. Pyramidal neurons in Kv1.3 KO mice exhibited significantly reduced paired-pulse ratio and increased presynaptic Cav2.1 expression, proving that the presynaptic vesicle release might be elevated by Ca2+ influx. Using Golgi staining, we found significantly increased dendritic spine density of pyramidal neurons in Kv1.3 KO mice, supporting the stronger postsynaptic responses in these neurons. In olfactory recognition and feeding behavior tests, we showed that Kv1.3 conditional knockout or cannula injection of 5-(4-phenoxybutoxy) psoralen, a Kv1.3 channel blocker, in piriform cortex both elevated the olfactory recognition index and altered the feeding behavior in mice. In summary, Kv1.3 is a key molecule in regulating neuronal activity of the piriform cortex, which may lay a foundation for the treatment of diseases related to piriform cortex and olfactory detection.
Collapse
Affiliation(s)
- Yong-Sheng Zhou
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, 430074, China
| | - Hao-Bo Tao
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, 430074, China
| | - Si-Si Lv
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, 430074, China
| | - Ke-Qin Liang
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, 430074, China
| | - Wen-Yi Shi
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, 430074, China
| | - Ke-Yi Liu
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, 430074, China
| | - Yun-Yun Li
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, 430074, China
| | - Lv-Yi Chen
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, 430074, China
| | - Ling Zhou
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, 430074, China
| | - Shi-Jin Yin
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, 430074, China.
| | - Qian-Ru Zhao
- Department of Chemical Biology, School of Pharmaceutical Sciences, South-Central Minzu University, Wuhan, 430074, China.
| |
Collapse
|
8
|
Xiong A, Richmond JE, Kim H. Presynaptic neurons self-tune by inversely coupling neurotransmitter release with the abundance of CaV2 voltage-gated Ca 2+ channels. Proc Natl Acad Sci U S A 2024; 121:e2404969121. [PMID: 39172783 PMCID: PMC11363341 DOI: 10.1073/pnas.2404969121] [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: 03/09/2024] [Accepted: 07/12/2024] [Indexed: 08/24/2024] Open
Abstract
The abundance of CaV2 voltage-gated calcium channels is linked to presynaptic homeostatic plasticity (PHP), a process that recalibrates synaptic strength to maintain the stability of neural circuits. However, the molecular and cellular mechanisms governing PHP and CaV2 channels are not completely understood. Here, we uncover a previously not described form of PHP in Caenorhabditis elegans, revealing an inverse regulatory relationship between the efficiency of neurotransmitter release and the abundance of UNC-2/CaV2 channels. Gain-of-function unc-2SL(S240L) mutants, which carry a mutation analogous to the one causing familial hemiplegic migraine type 1 in humans, showed markedly reduced channel abundance despite increased channel functionality. Reducing synaptic release in these unc-2SL(S240L) mutants restored channel levels to those observed in wild-type animals. Conversely, loss-of-function unc-2DA(D726A) mutants, which harbor the D726A mutation in the channel pore, exhibited a marked increase in channel abundance. Enhancing synaptic release in unc-2DA mutants reversed this increase in channel levels. Importantly, this homeostatic regulation of UNC-2 channel levels is accompanied by the structural remodeling of the active zone (AZ); specifically, unc-2DA mutants, which exhibit increased channel abundance, showed parallel increases in select AZ proteins. Finally, our forward genetic screen revealed that WWP-1, a HECT family E3 ubiquitin ligase, is a key homeostatic mediator that removes UNC-2 from synapses. These findings highlight a self-tuning PHP regulating UNC-2/CaV2 channel abundance along with AZ reorganization, ensuring synaptic strength and stability.
Collapse
Affiliation(s)
- Ame Xiong
- Discipline of Cell Biology & Anatomy, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL60064
| | - Janet E. Richmond
- Department of Biological Sciences, University of Illinois, Chicago, IL60607
| | - Hongkyun Kim
- Discipline of Cell Biology & Anatomy, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL60064
| |
Collapse
|
9
|
Chen C, Han P, Qing Y. Metabolic heterogeneity in tumor microenvironment - A novel landmark for immunotherapy. Autoimmun Rev 2024; 23:103579. [PMID: 39004158 DOI: 10.1016/j.autrev.2024.103579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/10/2024] [Accepted: 07/09/2024] [Indexed: 07/16/2024]
Abstract
The surrounding non-cancer cells and tumor cells that make up the tumor microenvironment (TME) have various metabolic rhythms. TME metabolic heterogeneity is influenced by the intricate network of metabolic control within and between cells. DNA, protein, transport, and microbial levels are important regulators of TME metabolic homeostasis. The effectiveness of immunotherapy is also closely correlated with alterations in TME metabolism. The response of a tumor patient to immunotherapy is influenced by a variety of variables, including intracellular metabolic reprogramming, metabolic interaction between cells, ecological changes within and between tumors, and general dietary preferences. Although immunotherapy and targeted therapy have made great strides, their use in the accurate identification and treatment of tumors still has several limitations. The function of TME metabolic heterogeneity in tumor immunotherapy is summarized in this article. It focuses on how metabolic heterogeneity develops and is regulated as a tumor progresses, the precise molecular mechanisms and potential clinical significance of imbalances in intracellular metabolic homeostasis and intercellular metabolic coupling and interaction, as well as the benefits and drawbacks of targeted metabolism used in conjunction with immunotherapy. This offers insightful knowledge and important implications for individualized tumor patient diagnosis and treatment plans in the future.
Collapse
Affiliation(s)
- Chen Chen
- The First Affiliated Hospital of Ningbo University, Ningbo 315211, Zhejiang, China
| | - Peng Han
- Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang, China.
| | - Yanping Qing
- The First Affiliated Hospital of Ningbo University, Ningbo 315211, Zhejiang, China.
| |
Collapse
|
10
|
Yang W, Chen C, Jiang X, Zhao Y, Wang J, Zhang Q, Zhang J, Feng Y, Cui S. CACNA1B protects naked mole-rat hippocampal neuron from apoptosis via altering the subcellular localization of Nrf2 after 60Co irradiation. Cell Biol Int 2024; 48:695-711. [PMID: 38389270 DOI: 10.1002/cbin.12140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 12/19/2023] [Accepted: 02/01/2024] [Indexed: 02/24/2024]
Abstract
Although radiotherapy is the most effective treatment modality for brain tumors, it always injures the central nervous system, leading to potential sequelae such as cognitive dysfunction. Radiation induces molecular, cellular, and functional changes in neuronal and glial cells. The hippocampus plays a critical role in learning and memory; therefore, concerns about radiation-induced injury are widespread. Multiple studies have focused on this complex problem, but the results have not been fully elucidated. Naked mole rat brains were irradiated with 60Co at a dose of 10 Gy. On 7 days, 14 days, and 28 days after irradiation, hippocampi in the control groups were obtained for next-generation sequencing. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were subsequently performed. Venn diagrams revealed 580 differentially expressed genes (DEGs) that were common at different times after irradiation. GO and KEGG analyses revealed that the 580 common DEGs were enriched in molecular transducer activity. In particular, CACNA1B mediated regulatory effects after irradiation. CACNA1B expression increased significantly after irradiation. Downregulation of CACNA1B led to a reduction in apoptosis and reactive oxygen species levels in hippocampal neurons. This was due to the interaction between CACNA1B and Nrf2, which disturbed the normal nuclear localization of Nrf2. In addition, CACNA1B downregulation led to a decrease in the cognitive functions of naked mole rats. These findings reveal the pivotal role of CACNA1B in regulating radiation-induced brain injury and will lead to the development of a novel strategy to prevent brain injury after irradiation.
Collapse
Affiliation(s)
- Wenjing Yang
- Laboratory Animal Science Department, Basic Medical School, Naval Medical University, Shanghai, China
| | - Chao Chen
- Laboratory Animal Science Department, Basic Medical School, Naval Medical University, Shanghai, China
| | - Xiaolong Jiang
- Laboratory Animal Science Department, Basic Medical School, Naval Medical University, Shanghai, China
| | - Yining Zhao
- Clinical Laboratory, Shanghai Yangpu district mental health center, Shanghai University of Medicine and Health Sciences Teaching Hospital, Shanghai, China
| | - Junyang Wang
- Laboratory Animal Science Department, Basic Medical School, Naval Medical University, Shanghai, China
| | - Qianqian Zhang
- Laboratory Animal Science Department, Basic Medical School, Naval Medical University, Shanghai, China
| | - Jingyuan Zhang
- Laboratory Animal Science Department, Basic Medical School, Naval Medical University, Shanghai, China
| | - Yan Feng
- Laboratory Animal Science Department, Basic Medical School, Naval Medical University, Shanghai, China
| | - Shufang Cui
- Laboratory Animal Science Department, Basic Medical School, Naval Medical University, Shanghai, China
| |
Collapse
|
11
|
Aldahabi M, Neher E, Nusser Z. Different states of synaptic vesicle priming explain target cell type-dependent differences in neurotransmitter release. Proc Natl Acad Sci U S A 2024; 121:e2322550121. [PMID: 38657053 PMCID: PMC11067035 DOI: 10.1073/pnas.2322550121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 03/27/2024] [Indexed: 04/26/2024] Open
Abstract
Pronounced differences in neurotransmitter release from a given presynaptic neuron, depending on the synaptic target, are among the most intriguing features of cortical networks. Hippocampal pyramidal cells (PCs) release glutamate with low probability to somatostatin expressing oriens-lacunosum-moleculare (O-LM) interneurons (INs), and the postsynaptic responses show robust short-term facilitation, whereas the release from the same presynaptic axons onto fast-spiking INs (FSINs) is ~10-fold higher and the excitatory postsynaptic currents (EPSCs) display depression. The mechanisms underlying these vastly different synaptic behaviors have not been conclusively identified. Here, we applied a combined functional, pharmacological, and modeling approach to address whether the main difference lies in the action potential-evoked fusion or else in upstream priming processes of synaptic vesicles (SVs). A sequential two-step SV priming model was fitted to the peak amplitudes of unitary EPSCs recorded in response to complex trains of presynaptic stimuli in acute hippocampal slices of adult mice. At PC-FSIN connections, the fusion probability (Pfusion) of well-primed SVs is 0.6, and 44% of docked SVs are in a fusion-competent state. At PC-O-LM synapses, Pfusion is only 40% lower (0.36), whereas the fraction of well-primed SVs is 6.5-fold smaller. Pharmacological enhancement of fusion by 4-AP and priming by PDBU was recaptured by the model with a selective increase of Pfusion and the fraction of well-primed SVs, respectively. Our results demonstrate that the low fidelity of transmission at PC-O-LM synapses can be explained by a low occupancy of the release sites by well-primed SVs.
Collapse
Affiliation(s)
- Mohammad Aldahabi
- Laboratory of Cellular Neurophysiology, Hungarian Research Network Institute of Experimental Medicine, Budapest1083, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University, Budapest1085, Hungary
| | - Erwin Neher
- Laboratory of Membrane Biophysics, Max Planck Institute for Multidisciplinary Sciences, 37077Göttingen, Germany
| | - Zoltan Nusser
- Laboratory of Cellular Neurophysiology, Hungarian Research Network Institute of Experimental Medicine, Budapest1083, Hungary
| |
Collapse
|
12
|
Li J, Veeraraghavan P, Young SM. Ca V 2.1 α 1 subunit motifs that control presynaptic Ca V 2.1 subtype abundance are distinct from Ca V 2.1 preference. J Physiol 2024; 602:485-506. [PMID: 38155373 PMCID: PMC10872416 DOI: 10.1113/jp284957] [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: 04/28/2023] [Accepted: 11/30/2023] [Indexed: 12/30/2023] Open
Abstract
Presynaptic voltage-gated Ca2+ channel (CaV ) subtype abundance at mammalian synapses regulates synaptic transmission in health and disease. In the mammalian central nervous system (CNS), most presynaptic terminals are CaV 2.1 dominant with a developmental reduction in CaV 2.2 and CaV 2.3 levels, and CaV 2 subtype levels are altered in various diseases. However, the molecular mechanisms controlling presynaptic CaV 2 subtype levels are largely unsolved. Because the CaV 2 α1 subunit cytoplasmic regions contain varying levels of sequence conservation, these regions are proposed to control presynaptic CaV 2 subtype preference and abundance. To investigate the potential role of these regions, we expressed chimeric CaV 2.1 α1 subunits containing swapped motifs with the CaV 2.2 and CaV 2.3 α1 subunit on a CaV 2.1/CaV 2.2 null background at the calyx of Held presynaptic terminals. We found that expression of CaV 2.1 α1 subunit chimeras containing the CaV 2.3 loop II-III region or cytoplasmic C-terminus (CT) resulted in a large reduction of presynaptic Ca2+ currents compared to the CaV 2.1 α1 subunit. However, the Ca2+ current sensitivity to the CaV 2.1 blocker agatoxin-IVA was the same between the chimeras and the CaV 2.1 α1 subunit. Additionally, we found no reduction in presynaptic Ca2+ currents with CaV 2.1/2.2 cytoplasmic CT chimeras. We conclude that the motifs in the CaV 2.1 loop II-III and CT do not individually regulate CaV 2.1 preference, although these motifs control CaV 2.1 levels and the CaV 2.3 CT contains motifs that negatively regulate presynaptic CaV 2.3 levels. We propose that the motifs controlling presynaptic CaV 2.1 preference are distinct from those regulating CaV 2.1 levels and may act synergistically to impact pathways regulating CaV 2.1 preference and abundance. KEY POINTS: Presynaptic CaV 2 subtype abundance regulates neuronal circuit properties, although the mechanisms regulating presynaptic CaV 2 subtype abundance and preference remain enigmatic. The CaV α1 subunit determines subtype and contains multiple motifs implicated in regulating presynaptic subtype abundance and preference. The CaV 2.1 α1 subunit domain II-III loop and cytoplasmic C-terminus are positive regulators of presynaptic CaV 2.1 abundance but do not regulate preference. The CaV 2.3 α1 subunit cytoplasmic C-terminus negatively regulates presynaptic CaV 2 subtype abundance but not preference, whereas the CaV 2.2 α1 subunit cytoplasmic C-terminus is not a key regulator of presynaptic CaV 2 subtype abundance or preference. The CaV 2 α1 subunit motifs determining the presynaptic CaV 2 preference are distinct from abundance.
Collapse
Affiliation(s)
- Jianing Li
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, USA
- Cell Developmental Biology Graduate Program, University of Iowa, Iowa City, IA 52242, USA
| | | | - Samuel M. Young
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, USA
- Department of Otolaryngology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
13
|
Li J, Veeraraghavan P, Young SM. CaV2.1 α1 subunit motifs that control presynaptic CaV2.1 subtype abundance are distinct from CaV2.1 preference. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.28.538778. [PMID: 37162941 PMCID: PMC10168310 DOI: 10.1101/2023.04.28.538778] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Presynaptic voltage-gated Ca2+ channels (CaV) subtype abundance at mammalian synapses regulates synaptic transmission in health and disease. In the mammalian central nervous system, most presynaptic terminals are CaV2.1 dominant with a developmental reduction in CaV2.2 and CaV2.3 levels, and CaV2 subtype levels are altered in various diseases. However, the molecular mechanisms controlling presynaptic CaV2 subtype levels are largely unsolved. Since the CaV2 α1 subunit cytoplasmic regions contain varying levels of sequence conservation, these regions are proposed to control presynaptic CaV2 subtype preference and abundance. To investigate the potential role of these regions, we expressed chimeric CaV2.1 α1subunits containing swapped motifs with the CaV2.2 and CaV2.3 α1 subunit on a CaV2.1/CaV2.2 null background at the calyx of Held presynaptic terminal. We found that expression of CaV2.1 α1 subunit chimeras containing the CaV2.3 loop II-III region or cytoplasmic C-terminus (CT) resulted in a large reduction of presynaptic Ca2+ currents compared to the CaV2.1 α1 subunit. However, the Ca2+ current sensitivity to the CaV2.1 blocker Agatoxin-IVA, was the same between the chimeras and the CaV2.1 α1 subunit. Additionally, we found no reduction in presynaptic Ca2+ currents with CaV2.1/2.2 cytoplasmic CT chimeras. We conclude that the motifs in the CaV2.1 loop II-III and CT do not individually regulate CaV2.1 preference, but these motifs control CaV2.1 levels and the CaV2.3 CT contains motifs that negatively regulate presynaptic CaV2.3 levels. We propose that the motifs controlling presynaptic CaV2.1 preference are distinct from those regulating CaV2.1 levels and may act synergistically to impact pathways regulating CaV2.1 preference and abundance.
Collapse
|
14
|
Keine C, Al-Yaari M, Radulovic T, Young SM. Stereotactic Delivery of Helper-dependent Adenoviral Viral Vectors at Distinct Developmental Time Points to Perform Age-dependent Molecular Manipulations of the Mouse Calyx of Held. Bio Protoc 2023; 13:e4793. [PMID: 37638292 PMCID: PMC10450731 DOI: 10.21769/bioprotoc.4793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/23/2023] [Accepted: 06/24/2023] [Indexed: 08/29/2023] Open
Abstract
Synapses are specialized structures that enable neuronal communication, which is essential for brain function and development. Alterations in synaptic proteins have been linked to various neurological and neuropsychiatric disorders. Therefore, manipulating synaptic proteins in vivo can provide insight into the molecular mechanisms underlying these disorders and aid in developing new therapeutic strategies. Previous methods such as constitutive knock-out animals are limited by developmental compensation and off-target effects. The current approach outlines procedures for age-dependent molecular manipulations in mice using helper-dependent adenovirus viral vectors (HdAd) at distinct developmental time points. Using stereotactic injection of HdAds in both newborn and juvenile mice, we demonstrate the versatility of this method to express Cre recombinase in globular bushy cells of juvenile Rac1fl/fl mice to ablate presynaptic Rac1 and study its role in synaptic transmission. Separately, we overexpress CaV2 α1 subunits at two distinct developmental time points to elucidate the mechanisms that determine presynaptic CaV2 channel abundance and preference. This method presents a reliable, cost-effective, and minimally invasive approach for controlling gene expression in specific regions of the mouse brain and will be a powerful tool to decipher brain function in health and disease. Key features Virus-mediated genetic perturbation in neonatal and young adult mice. Stereotaxic injection allows targeting of brain structures at different developmental stages to study the impact of genetic perturbation throughout the development.
Collapse
Affiliation(s)
- Christian Keine
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, USA
- Department of Human Medicine, University of Oldenburg, Oldenburg, Germany
- Research Center Neurosensory Science, Oldenburg, Germany
| | - Mohammed Al-Yaari
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, USA
| | - Tamara Radulovic
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, USA
- Department of Human Medicine, University of Oldenburg, Oldenburg, Germany
- Research Center Neurosensory Science, Oldenburg, Germany
| | - Samuel M. Young
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, USA
- Department of Otolaryngology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
15
|
Kramer AA, Bennett DF, Barañano KW, Bannister RA. A neurodevelopmental disorder caused by a dysfunctional CACNA1A allele. eNeurologicalSci 2023; 31:100456. [PMID: 36938367 PMCID: PMC10020665 DOI: 10.1016/j.ensci.2023.100456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/05/2022] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
P/Q-type Ca2+ flux into nerve terminals via CaV2.1 channels is essential for neurotransmitter release at neuromuscular junctions and nearly all central synapses. Mutations in CACNA1A, the gene encoding CaV2.1, cause a spectrum of pediatric neurological disorders. We have identified a patient harboring an autosomal-dominant de novo frameshift-causing nucleotide duplication in CACNA1A (c.5018dupG). The duplicated guanine precipitated 43 residues of altered amino acid sequence beginning with a glutamine to serine substitution in CaV2.1 at position 1674 ending with a premature stop codon (CaV2.1 p.Gln1674Serfs*43). The patient presented with episodic downbeat vertical nystagmus, hypotonia, ataxia, developmental delay and febrile seizures. In patch-clamp experiments, no Ba2+ current was observed in tsA-201 cells expressing CaV2.1 p.Gln1674Serfs*43 with β4 and α2δ-1 auxiliary subunits. The ablation of divalent flux in response to depolarization was likely attributable to the inability of CaV2.1 p.Gln1674Serfs*43 to form a complete channel pore. Our results suggest that the pathology resulting from this frameshift-inducing nucleotide duplication is a consequence of an effective haploinsufficiency.
Collapse
Affiliation(s)
- Audra A. Kramer
- Department of Pathology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 North Greene Street, Baltimore, MD 21201, USA
- Corresponding authors at: National Institutes of Health, Center for Scientific Review, Division of Neuroscience, Development and Aging, 6701 Rockledge Drive, Bethesda, MD 20892, USA.
| | - Daniel F. Bennett
- Department of Pathology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 North Greene Street, Baltimore, MD 21201, USA
| | - Kristin W. Barañano
- Department of Neurology, Johns Hopkins University School of Medicine, 200 North Wolfe Street, Suite 2158, Baltimore, MD 21287, USA
| | - Roger A. Bannister
- Department of Pathology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 North Greene Street, Baltimore, MD 21201, USA
- Corresponding authors at: National Institutes of Health, Center for Scientific Review, Division of Neuroscience, Development and Aging, 6701 Rockledge Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
16
|
Chen X, Chen S, Li Z, Zhu R, Jia Z, Ban J, Zhen R, Chen X, Pan X, Ren Q, Yue L, Niu S. Effect of semaglutide and empagliflozin on cognitive function and hippocampal phosphoproteomic in obese mice. Front Pharmacol 2023; 14:975830. [PMID: 37007007 PMCID: PMC10063902 DOI: 10.3389/fphar.2023.975830] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 03/03/2023] [Indexed: 03/19/2023] Open
Abstract
Objective: Based on the 4D label-free phosphoproteomic technique, we examined the differences in cognitive function and hippocampal phosphorylated protein expression in high-fat diet-induced obese mice after the intervention of semaglutide and empagliflozin, as well as the effects of both on protein activity and function in obese mice’s hippocampal tissues and the signaling pathways involved.Methods: Thirty-two C57BL/6JC male mice were assigned to two groups randomly: A control group (group C, 10% of energy is from fat, n = 8) and a high-fat diet group (group H, 60% of energy is from fat, n = 24). The high-fat diet-induced obese mice were screened after 12 weeks of feeding based on the criterion that the bodyweight of mice in fat rich diet group was greater than or equal to 20% of the average body weight of the mice in the blank control group. Group H separate into group H (n = 8), group Semaglutide (group S, n = 8), and group empagliflozin (group E, n = 8). For a total of 12 weeks, group S received 30 nmol/kg/d bodyweight of semaglutide intraperitoneally, group E received 10 mg/kg/d bodyweight of empagliflozin via gavage, and groups C and H received equal amounts of saline by intraperitoneal injection and gavage. At the end of treatment, the mice were appraised for cognitive function employing the Morris water maze (MWM), and serum fasting glucose, lipids, and inflammatory parameters were measured. The 4D label-free phosphoproteomics method was employed to screen the differential phosphoproteins and loci in hippocampal tissues of mice in different treatment groups, and bioinformatics was used to analyze the biological processes, signaling pathways, and related protein–protein interaction (PPI) network analysis of these differentially phosphorylated proteins.Results: In comparison to normal controls, The escape latency of obese mice induced by high-fat diet was prolonged, the percentage of swimming time in the target quadrant was reduced, and the number of times of crossing the platform was reduced, whereas semaglutide and empagliflozin treatment reduced escape latency, increase the percentage of swim time in the target quadrant and increase the frequency of passing through the platform area, although there is little difference in the effect of the two drugs. The phosphoproteomic results showed 20,493 unique phosphorylated peptides, representing 21,239 phosphorylation sites and 4,290 phosphorylated proteins. Further analysis revealed that the proteins corresponding to these differentially phosphorylated sites are jointly distributed in signaling pathways such as dopaminergic synapses and axon guidance, and are involved in biological processes such as neuronal projection development, synaptic plasticity, and axonogenesis. Notably, the key factors voltage-dependent L-type calcium channel subunit alpha-1D (CACNA1D), voltage-dependent P/Q-type calcium channel subunit alpha-1A (CACNA1A), and voltage-dependent N-type calcium channel subunit alpha-1B (CACNA1B) were all found to be involved in the dopaminergic synapse pathway, and their expression was upregulated by semaglutide and empagliflozin.Conclusion: We found for the first time that a high-fat diet decreased CACNA1D, CACNA1A, and CACNA1B protein serine phosphorylation, which may affect neuronal development, synaptic plasticity, and cognitive function in mice. Notably, semaglutide and empagliflozin increased the phosphorylation of these proteins.
Collapse
Affiliation(s)
- Xiaoyi Chen
- Department of Internal Medicine, Hebei North University, Zhangjiakou, China
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
| | - Shuchun Chen
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
- *Correspondence: Shuchun Chen,
| | - Zelin Li
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
| | - Ruiyi Zhu
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
| | - Zhuoya Jia
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
| | - Jiangli Ban
- Department of Internal Medicine, Hebei North University, Zhangjiakou, China
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
| | - Ruoxi Zhen
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
| | - Xing Chen
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
| | - Xiaoyu Pan
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
| | - Qingjuan Ren
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
| | - Lin Yue
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
| | - Shu Niu
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, China
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
17
|
Cunningham KL, Littleton JT. Mechanisms controlling the trafficking, localization, and abundance of presynaptic Ca 2+ channels. Front Mol Neurosci 2023; 15:1116729. [PMID: 36710932 PMCID: PMC9880069 DOI: 10.3389/fnmol.2022.1116729] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 12/26/2022] [Indexed: 01/14/2023] Open
Abstract
Voltage-gated Ca2+ channels (VGCCs) mediate Ca2+ influx to trigger neurotransmitter release at specialized presynaptic sites termed active zones (AZs). The abundance of VGCCs at AZs regulates neurotransmitter release probability (Pr ), a key presynaptic determinant of synaptic strength. Given this functional significance, defining the processes that cooperate to establish AZ VGCC abundance is critical for understanding how these mechanisms set synaptic strength and how they might be regulated to control presynaptic plasticity. VGCC abundance at AZs involves multiple steps, including channel biosynthesis (transcription, translation, and trafficking through the endomembrane system), forward axonal trafficking and delivery to synaptic terminals, incorporation and retention at presynaptic sites, and protein recycling. Here we discuss mechanisms that control VGCC abundance at synapses, highlighting findings from invertebrate and vertebrate models.
Collapse
Affiliation(s)
- Karen L. Cunningham
- The Picower Institute for Learning and Memory, Department of Biology, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| | | |
Collapse
|
18
|
Aldahabi M, Balint F, Holderith N, Lorincz A, Reva M, Nusser Z. Different priming states of synaptic vesicles underlie distinct release probabilities at hippocampal excitatory synapses. Neuron 2022; 110:4144-4161.e7. [PMID: 36261033 PMCID: PMC9796815 DOI: 10.1016/j.neuron.2022.09.035] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/28/2022] [Accepted: 09/27/2022] [Indexed: 12/12/2022]
Abstract
A stunning example of synaptic diversity is the postsynaptic target cell-type-dependent difference in synaptic efficacy in cortical networks. Here, we show that CA1 pyramidal cell (PC) to fast spiking interneuron (FSIN) connections have 10-fold larger release probability (Pv) than those on oriens lacunosum-moleculare (O-LM) interneurons. Freeze-fracture immunolabeling revealed that different nano-topologies and coupling distances between Ca2+ channels and release sites (RSs) are not responsible for the distinct Pv. Although [Ca2+] transients are 40% larger in FSINs innervating boutons, when [Ca2+] entry is matched in the two bouton populations, EPSCs in O-LM cells are still 7-fold smaller. However, application of a phorbol ester analog resulted in a ∼2.5-fold larger augmentation at PC - O-LM compared to PC - FSIN synapses, suggesting incomplete docking or priming of vesicles. Similar densities of docked vesicles rule out distinct RS occupancies and demonstrate that incompletely primed, but docked, vesicles limit the output of PC - O-LM synapses.
Collapse
Affiliation(s)
- Mohammad Aldahabi
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Budapest 1083, Hungary,János Szentágothai School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Flora Balint
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Noemi Holderith
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Andrea Lorincz
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Maria Reva
- Unit of Synapse and Circuit Dynamics, CNRS UMR 3571, Institute Pasteur, Paris, France
| | - Zoltan Nusser
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Budapest 1083, Hungary,Corresponding author
| |
Collapse
|
19
|
Zhang W, Jiang HH, Luo F. Diverse organization of voltage-gated calcium channels at presynaptic active zones. Front Synaptic Neurosci 2022; 14:1023256. [PMID: 36544543 PMCID: PMC9760684 DOI: 10.3389/fnsyn.2022.1023256] [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: 08/19/2022] [Accepted: 11/09/2022] [Indexed: 12/12/2022] Open
Abstract
Synapses are highly organized but are also highly diverse in their organization and properties to allow for optimizing the computing power of brain circuits. Along these lines, voltage-gated calcium (CaV) channels at the presynaptic active zone are heterogeneously organized, which creates a variety of calcium dynamics profiles that can shape neurotransmitter release properties of individual synapses. Extensive studies have revealed striking diversity in the subtype, number, and distribution of CaV channels, as well as the nanoscale topographic relationships to docked synaptic vesicles. Further, multi-protein complexes including RIMs, RIM-binding proteins, CAST/ELKS, and neurexins are required for coordinating the diverse organization of CaV channels at the presynaptic active zone. In this review, we highlight major advances in the studies of the functional organization of presynaptic CaV channels and discuss their physiological implications for synaptic transmission and short-term plasticity.
Collapse
Affiliation(s)
- Weijia Zhang
- Guangzhou Laboratory, Guangzhou, China,Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - He-Hai Jiang
- Guangzhou Laboratory, Guangzhou, China,Bioland Laboratory, Guangzhou, China
| | - Fujun Luo
- Guangzhou Laboratory, Guangzhou, China,Bioland Laboratory, Guangzhou, China,*Correspondence: Fujun Luo
| |
Collapse
|
20
|
Bahena-Alvarez D, Millan-Aldaco D, Rincón-Heredia R, Escamilla-Avila N, Hernandez-Cruz A. Expression of voltage-gated Ca2+ channels, Insp3Rs, and RyRs in the immature mouse ovary. J Ovarian Res 2022; 15:85. [PMID: 35869556 PMCID: PMC9306205 DOI: 10.1186/s13048-022-01015-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 06/27/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The postnatal mammalian ovary undergoes a series of changes to ensure the maturation of sufficient follicles to support ovulation and fecundation over the reproductive life. It is well known that intracellular [Ca2+]i signals are necessary for ovulation, fertilization, and egg activation. However, we lack detailed knowledge of the molecular identity, cellular distribution, and functional role of Ca2+ channels expressed during folliculogenesis. In the neonatal period, ovarian maturation is controlled by protein growth factors released from the oocyte and granulosa cells. Conversely, during the early infantile period, maturation becomes gonadotropin-dependent and is controlled by granulosa and theca cells. The significance of intracellular Ca2+ signaling in folliculogenesis is supported by the observation that mice lacking the expression of Ca2+/calmodulin-dependent kinase IV in granulosa cells suffer abnormal follicular development and impaired fertility.
Results
Using immunofluorescence in frozen ovarian sections and confocal microscopy, we assessed the expression of high-voltage activated Ca2+ channel alpha subunits and InsP3 and ryanodine receptors in the postnatal period from 3 to 16 days. During the neonatal stage, oocytes from primordial and primary follicles show high expression of various Ca2+-selective channels, with granulosa and stroma cells expressing significantly less. These channels are likely involved in supporting Ca2+-dependent secretion of peptide growth factors. In contrast, during the early and late infantile periods, Ca2+ channel expression in the oocyte diminishes, increasing significantly in the granulosa and particularly in immature theca cells surrounding secondary follicles.
Conclusions
The developmental switch of Ca2+ channel expression from the oocytes to the perifollicular cells likely reflects the vanishing role of the oocytes once granulosa and theca cells take control of folliculogenesis in response to gonadotropins acting on their receptors.
Collapse
|
21
|
Midorikawa M. Developmental and activity-dependent modulation of coupling distance between release site and Ca2+ channel. Front Cell Neurosci 2022; 16:1037721. [DOI: 10.3389/fncel.2022.1037721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/11/2022] [Indexed: 11/13/2022] Open
Abstract
Synapses are junctions between a presynaptic neuron and a postsynaptic cell specialized for fast and precise information transfer. The presynaptic terminal secretes neurotransmitters via exocytosis of synaptic vesicles. Exocytosis is a tightly regulated reaction that occurs within a millisecond of the arrival of an action potential. One crucial parameter in determining the characteristics of the transmitter release kinetics is the coupling distance between the release site and the Ca2+ channel. Still, the technical limitations have hindered detailed analysis from addressing how the coupling distance is regulated depending on the development or activity of the synapse. However, recent technical advances in electrophysiology and imaging are unveiling their different configurations in different conditions. Here, I will summarize developmental- and activity-dependent changes in the coupling distances revealed by recent studies.
Collapse
|
22
|
Midorikawa M. Pathway-specific maturation of presynaptic functions of the somatosensory thalamus. Neurosci Res 2022; 181:1-8. [DOI: 10.1016/j.neures.2022.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 02/05/2023]
|
23
|
Guerrero-Given D, Goldin SL, Thomas CI, Anthony SA, Jerez D, Kamasawa N. Gold In-and-Out: A Toolkit for Analyzing Subcellular Distribution of Immunogold-Labeled Membrane Proteins in Freeze-Fracture Replica Images. Front Neuroanat 2022; 16:855218. [PMID: 35444519 PMCID: PMC9014018 DOI: 10.3389/fnana.2022.855218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/04/2022] [Indexed: 11/13/2022] Open
Abstract
Integral membrane proteins such as ion channels, transporters, and receptors shape cell activity and mediate cell-to-cell communication in the brain. The distribution, quantity, and clustering arrangement of those proteins contribute to the physiological properties of the cell; therefore, precise quantification of their state can be used to gain insight into cellular function. Using a highly sensitive immunoelectron microscopy technique called sodium dodecyl sulfate-digested freeze-fracture replica immunogold labeling (SDS-FRL), multiple membrane proteins can be tagged with different sizes of immunogold particles at once and visualized two-dimensionally. For quantification, gold particles in the images must be annotated, and then different mathematical and statistical methods must be applied to characterize the distribution states of proteins of interest. To perform such analyses in a user-friendly manner, we developed a program with a simple graphical user interface called Gold In-and-Out (GIO), which integrates several classical and novel analysis methods for immunogold labeled replicas into one self-contained package. GIO takes an input of particle coordinates, then allows users to implement analysis methods such as nearest neighbor distance (NND) and particle clustering. The program not only performs the selected analysis but also automatically compares the results of the real distribution to a random distribution of the same number of particles on the membrane region of interest. In addition to classical approaches for analyzing protein distribution, GIO includes new tools to analyze the positional bias of a target protein relative to a morphological landmark such as dendritic spines, and can also be applied for synaptic protein analysis. Gold Rippler provides a normalized metric of particle density that is resistant to differences in labeling efficiency among samples, while Gold Star is useful for quantifying distances between a protein and landmark. This package aims to help standardize analysis methods for subcellular and synaptic protein localization with a user-friendly interface while increasing the efficiency of these time-consuming analyses.
Collapse
Affiliation(s)
| | | | | | | | | | - Naomi Kamasawa
- The Imaging Center and Electron Microscopy Core Facility, Max Planck Institute for Neuroscience, Jupiter, FL, United States
| |
Collapse
|
24
|
Stahl A, Noyes NC, Boto T, Botero V, Broyles CN, Jing M, Zeng J, King LB, Li Y, Davis RL, Tomchik SM. Associative learning drives longitudinally graded presynaptic plasticity of neurotransmitter release along axonal compartments. eLife 2022; 11:e76712. [PMID: 35285796 PMCID: PMC8956283 DOI: 10.7554/elife.76712] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/11/2022] [Indexed: 12/27/2022] Open
Abstract
Anatomical and physiological compartmentalization of neurons is a mechanism to increase the computational capacity of a circuit, and a major question is what role axonal compartmentalization plays. Axonal compartmentalization may enable localized, presynaptic plasticity to alter neuronal output in a flexible, experience-dependent manner. Here, we show that olfactory learning generates compartmentalized, bidirectional plasticity of acetylcholine release that varies across the longitudinal compartments of Drosophila mushroom body (MB) axons. The directionality of the learning-induced plasticity depends on the valence of the learning event (aversive vs. appetitive), varies linearly across proximal to distal compartments following appetitive conditioning, and correlates with learning-induced changes in downstream mushroom body output neurons (MBONs) that modulate behavioral action selection. Potentiation of acetylcholine release was dependent on the CaV2.1 calcium channel subunit cacophony. In addition, contrast between the positive conditioned stimulus and other odors required the inositol triphosphate receptor, which maintained responsivity to odors upon repeated presentations, preventing adaptation. Downstream from the MB, a set of MBONs that receive their input from the γ3 MB compartment were required for normal appetitive learning, suggesting that they represent a key node through which reward learning influences decision-making. These data demonstrate that learning drives valence-correlated, compartmentalized, bidirectional potentiation, and depression of synaptic neurotransmitter release, which rely on distinct mechanisms and are distributed across axonal compartments in a learning circuit.
Collapse
Affiliation(s)
- Aaron Stahl
- Department of Neuroscience, The Scripps Research InstituteJupiterUnited States
| | - Nathaniel C Noyes
- Department of Neuroscience, The Scripps Research InstituteJupiterUnited States
| | - Tamara Boto
- Department of Neuroscience, The Scripps Research InstituteJupiterUnited States
| | - Valentina Botero
- Department of Neuroscience, The Scripps Research InstituteJupiterUnited States
| | - Connor N Broyles
- Department of Neuroscience, The Scripps Research InstituteJupiterUnited States
| | - Miao Jing
- Chinese Institute for Brain ResearchBeijingChina
| | - Jianzhi Zeng
- Peking-Tsinghua Center for Life Sciences, Peking UniversityBeijingChina
- State Key Laboratory of Membrane Biology, Peking University School of Life SciencesBeijingChina
- PKU IDG/McGovern Institute for Brain ResearchBeijingChina
| | - Lanikea B King
- Department of Neuroscience, The Scripps Research InstituteJupiterUnited States
| | - Yulong Li
- Chinese Institute for Brain ResearchBeijingChina
- Peking-Tsinghua Center for Life Sciences, Peking UniversityBeijingChina
- State Key Laboratory of Membrane Biology, Peking University School of Life SciencesBeijingChina
- PKU IDG/McGovern Institute for Brain ResearchBeijingChina
| | - Ronald L Davis
- Department of Neuroscience, The Scripps Research InstituteJupiterUnited States
| | - Seth M Tomchik
- Department of Neuroscience, The Scripps Research InstituteJupiterUnited States
| |
Collapse
|
25
|
Phillips S, Ramos PV, Veeraraghavan P, Young SM. VikAD, a Vika site-specific recombinase-based system for efficient and scalable helper-dependent adenovirus production. Mol Ther Methods Clin Dev 2022; 24:117-126. [PMID: 35024378 PMCID: PMC8718833 DOI: 10.1016/j.omtm.2021.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 12/04/2021] [Indexed: 11/09/2022]
Abstract
Recombinant viral vectors have become integral tools for basic in vivo research applications. Helper-dependent adenoviral (HdAd) vectors have a large packaging capacity of ∼36 kb of DNA that mediate long-term transgene expression in vitro and in vivo. The large carrying capacity of HdAd enables basic research or clinical applications requiring the delivery of large genes or multiple transgenes, which cannot be packaged into other widely used viral vectors. Currently, common HdAd production systems use an Ad helper virus (HV) with a packaging signal (Ψ) that is flanked by either loxP or FRT sites, which is excised in producer cells expressing Cre or Flp recombinases to prevent HV packaging. However, these production systems prevent the use of HdAd vectors for genetic strategies that rely on Cre or Flp recombination for cell-type-specific expression. To overcome these limitations, we developed the VikAD production system, which is based on producer cells expressing the Vika recombinase and an HV that contains a Ψ flanked by vox sites. The availability of this production system will greatly expand the utility and flexibility of HdAd vectors for use in research applications to monitor and manipulate cellular activity with increased specificity.
Collapse
Affiliation(s)
- Stacia Phillips
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, PBDB 5322, 169 Newton Road, Iowa City, IA 52242, USA
| | - Paula Valino Ramos
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, PBDB 5322, 169 Newton Road, Iowa City, IA 52242, USA
| | - Priyadharishini Veeraraghavan
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, PBDB 5322, 169 Newton Road, Iowa City, IA 52242, USA
| | - Samuel M. Young
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, PBDB 5322, 169 Newton Road, Iowa City, IA 52242, USA
- Department of Otolaryngology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| |
Collapse
|
26
|
The complexities of CACNA1A in clinical neurogenetics. J Neurol 2021; 269:3094-3108. [PMID: 34806130 DOI: 10.1007/s00415-021-10897-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 11/03/2021] [Accepted: 11/05/2021] [Indexed: 12/25/2022]
Abstract
Variants in CACNA1A are classically related to episodic ataxia type 2, familial hemiplegic migraine type 1, and spinocerebellar ataxia type 6. Over the years, CACNA1A has been associated with a broader spectrum of phenotypes. Targeted analysis and unbiased sequencing of CACNA1A result not only in clear molecular diagnoses, but also in large numbers of variants of uncertain significance (VUS), or likely pathogenic variants with a phenotype that does not directly match the CACNA1A spectrum. Over the last years, targeted and clinical exome sequencing in our center has identified 41 CACNA1A variants. Ultimately, variants were considered pathogenic or likely pathogenic in 23 cases, with most phenotypes ranging from episodic or progressive ataxia to more complex ataxia syndromes, as well as intellectual disability and epilepsy. In two cases, the causality of the variant was discarded based on non-segregation or an alternative diagnosis. In the remaining 16 cases, the variant was classified as uncertain, due to lack of opportunities for segregation analysis or uncertain association with a non-classic phenotype. Phenotypic variability and the large number of VUS make CACNA1A a challenging gene for neurogenetic diagnostics. Accessible functional read-outs are clearly needed, especially in cases with a non-classic phenotype.
Collapse
|
27
|
Yang CH, Ho WK, Lee SH. Postnatal maturation of glutamate clearance and release kinetics at the rat and mouse calyx of Held synapses. Synapse 2021; 75:e22215. [PMID: 34057239 DOI: 10.1002/syn.22215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 05/04/2021] [Accepted: 05/23/2021] [Indexed: 11/09/2022]
Abstract
Although calyx of Held synapses undergo dramatic changes around the hearing onset, previous in vivo studies suggest that the calyx synapses undergo further post-hearing maturation process. While developmental changes over the hearing onset have been extensively studied, this post-hearing maturation process remained relatively little investigated. Because of post-hearing maturation, previous results from studies around hearing onset and studies of post-hearing calyx synapses are somewhat inconsistent. Here, we characterized the post-hearing maturation of calyx synapses with regard to in vitro electrophysiological properties in rats and mice. We found that parameters for residual glutamate in the cleft during a train, EPSC kinetics, and vesicle pool size became close to a full mature level by P14, but they further matured until P16 in the rats. Consistently, the phasic and slow EPSCs evoked by action potential trains at P16 calyx synapses were not different from those at P18 or P25 under physiological extracellular [Ca2+ ]o (1.2 mM). In contrast, the parameters for residual current and EPSC kinetics displayed drastic changes until P16 in mice, and slow EPSCs during the train further decreased between P16 and P18, suggesting that maturation of calyx synapses progresses at least up to P16 in rats and P18 in mice.
Collapse
Affiliation(s)
- Che Ho Yang
- Cell Physiology Lab, Department of Physiology, Seoul National University College of Medicine and Neuroscience Research Institute, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Won-Kyung Ho
- Cell Physiology Lab, Department of Physiology, Seoul National University College of Medicine and Neuroscience Research Institute, Seoul National University Medical Research Center, Seoul, Republic of Korea.,Department of Brain and Cognitive Science, Seoul National University, Seoul, Republic of Korea
| | - Suk-Ho Lee
- Cell Physiology Lab, Department of Physiology, Seoul National University College of Medicine and Neuroscience Research Institute, Seoul National University Medical Research Center, Seoul, Republic of Korea.,Department of Brain and Cognitive Science, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
28
|
Oh KH, Krout MD, Richmond JE, Kim H. UNC-2 CaV2 Channel Localization at Presynaptic Active Zones Depends on UNC-10/RIM and SYD-2/Liprin-α in Caenorhabditis elegans. J Neurosci 2021; 41:4782-4794. [PMID: 33975919 PMCID: PMC8260173 DOI: 10.1523/jneurosci.0076-21.2021] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 04/07/2021] [Accepted: 04/29/2021] [Indexed: 12/20/2022] Open
Abstract
Presynaptic active zone proteins couple calcium influx with synaptic vesicle exocytosis. However, the control of presynaptic calcium channel localization by active zone proteins is not completely understood. In a Caenorhabditis elegans (C. elegans) forward genetic screen, we find that UNC-10/RIM (Rab3-interacting molecule) and SYD-2/Liprin-α regulate presynaptic localization of UNC-2, the CaV2 channel ortholog. We further quantitatively analyzed live animals using endogenously GFP-tagged UNC-2 and active zone components. Consistent with the interaction between RIM and CaV2 in mammals, the intensity and number of UNC-2 channel puncta at presynaptic terminals were greatly reduced in unc-10 mutant animals. To understand how SYD-2 regulates presynaptic UNC-2 channel localization, we analyzed presynaptic localization of endogenous SYD-2, UNC-10, RIMB-1/RIM-BP (RIM binding protein), and ELKS-1. Our analysis revealed that although SYD-2 is the most critical for active zone assembly, loss of SYD-2 function does not completely abolish presynaptic localization of UNC-10, RIMB-1, and ELKS-1, suggesting an existence of SYD-2-independent active zone assembly. UNC-2 localization analysis in double and triple mutants of active zone components show that SYD-2 promotes UNC-2 localization by partially controlling UNC-10 localization, and ELKS-1 and RIMB-1 also contribute to UNC-2 channel localization. In addition, we find that core active zone proteins are unequal in their abundance. Although the abundance of UNC-10 at the active zone is comparable to UNC-2, SYD-2 and ELKS-1 are twice more and RIMB-1 four times more abundant than UNC-2. Together our data show that UNC-10, SYD-2, RIMB-1, and ELKS-1 control presynaptic UNC-2 channel localization in redundant yet distinct manners.SIGNIFICANCE STATEMENT Precise control of neurotransmission is dependent on the tight coupling of the calcium influx through voltage-gated calcium channels (VGCCs) to the exocytosis machinery at the presynaptic active zones. However, how these VGCCs are tethered to the active zone is incompletely understood. To understand the mechanism of presynaptic VGCC localization, we performed a C. elegans forward genetic screen and quantitatively analyzed endogenous active zones and presynaptic VGCCs. In addition to RIM, our study finds that SYD-2/Liprin-α is critical for presynaptic localization of VGCCs. Yet, the loss of SYD-2, a core active zone scaffolding protein, does not completely abolish the presynaptic localization of the VGCC, showing that the active zone is a resilient structure assembled by redundant mechanisms.
Collapse
Affiliation(s)
- Kelly H Oh
- Center for Cancer Cell Biology, Immunology, and Infection, Department of Cell Biology and Anatomy, Chicago Medical School, School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois 60064
| | - Mia D Krout
- Department of Biological Science, University of Illinois at Chicago, Chicago, Illinois 60607
| | - Janet E Richmond
- Department of Biological Science, University of Illinois at Chicago, Chicago, Illinois 60607
| | - Hongkyun Kim
- Center for Cancer Cell Biology, Immunology, and Infection, Department of Cell Biology and Anatomy, Chicago Medical School, School of Graduate and Postdoctoral Studies, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois 60064
| |
Collapse
|
29
|
Environmental Enrichment Enhances Ca v 2.1 Channel-Mediated Presynaptic Plasticity in Hypoxic-Ischemic Encephalopathy. Int J Mol Sci 2021; 22:ijms22073414. [PMID: 33810296 PMCID: PMC8037860 DOI: 10.3390/ijms22073414] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 03/23/2021] [Indexed: 12/16/2022] Open
Abstract
Hypoxic–ischemic encephalopathy (HIE) is a devastating neonatal brain condition caused by lack of oxygen and limited blood flow. Environmental enrichment (EE) is a classic paradigm with a complex stimulation of physical, cognitive, and social components. EE can exert neuroplasticity and neuroprotective effects in immature brains. However, the exact mechanism of EE on the chronic condition of HIE remains unclear. HIE was induced by a permanent ligation of the right carotid artery, followed by an 8% O2 hypoxic condition for 1 h. At 6 weeks of age, HIE mice were randomly assigned to either standard cages or EE cages. In the behavioral assessments, EE mice showed significantly improved motor performances in rotarod tests, ladder walking tests, and hanging wire tests, compared with HIE control mice. EE mice also significantly enhanced cognitive performances in Y-maze tests. Particularly, EE mice showed a significant increase in Cav 2.1 (P/Q type) and presynaptic proteins by molecular assessments, and a significant increase of Cav 2.1 in histological assessments of the cerebral cortex and hippocampus. These results indicate that EE can upregulate the expression of the Cav 2.1 channel and presynaptic proteins related to the synaptic vesicle cycle and neurotransmitter release, which may be responsible for motor and cognitive improvements in HIE.
Collapse
|
30
|
Distinct functional developments of surviving and eliminated presynaptic terminals. Proc Natl Acad Sci U S A 2021; 118:2022423118. [PMID: 33688051 DOI: 10.1073/pnas.2022423118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
For neuronal circuits in the brain to mature, necessary synapses must be maintained and redundant synapses eliminated through experience-dependent mechanisms. However, the functional differentiation of these synapse types during the refinement process remains elusive. Here, we addressed this issue by distinct labeling and direct recordings of presynaptic terminals fated for survival and for elimination in the somatosensory thalamus. At surviving terminals, the number of total releasable vesicles was first enlarged, and then calcium channels and fast-releasing synaptic vesicles were tightly coupled in an experience-dependent manner. By contrast, transmitter release mechanisms did not mature at terminals fated for elimination, irrespective of sensory experience. Nonetheless, terminals fated for survival and for elimination both exhibited developmental shortening of action potential waveforms that was experience independent. Thus, we dissected experience-dependent and -independent developmental maturation processes of surviving and eliminated presynaptic terminals during neuronal circuit refinement.
Collapse
|
31
|
Prakash C, Mishra M, Kumar P, Kumar V, Sharma D. Response of Voltage-Gated Sodium and Calcium Channels Subtypes on Dehydroepiandrosterone Treatment in Iron-Induced Epilepsy. Cell Mol Neurobiol 2021; 41:279-292. [PMID: 32318899 PMCID: PMC11448620 DOI: 10.1007/s10571-020-00851-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/14/2020] [Indexed: 12/28/2022]
Abstract
Epilepsy is a neurological disorder characterized by the occurrence of spontaneous and recurrent seizures. In post-traumatic epilepsy (PTE), the mechanism of epileptogenesis is very complex and seems to be linked with voltage-gated ion channels. Dehydroepiandrosterone (DHEA), a neurosteroid have shown beneficial effect against various neurological disorders. We investigated antiepileptic effect of DHEA with respect to expression of voltage-gated ion channels subtypes in iron-induced epilepsy. Iron (FeCl3) solution was intracartically injected to induce epilepsy in rats and DHEA was intraperitoneally administered for 21 days. Results showed markedly increased epileptiform seizures activity along with up-regulation of Nav1.1 and Nav1.6, and down-regulation of Cav2.1α at the mRNA and protein level in the cortex and hippocampus of epileptic rats. Moreover, the study demonstrated that these channels subtypes were predominantly expressed in the neurons. DHEA treatment has countered the epileptic seizures, down-regulated Nav1.1 and Nav1.6, and up-regulated Cav2.1α without affecting their cellular localization. In conclusion, the present study demonstrates antiepileptic potential of DHEA, escorted by regulation of Nav1.1, Nav1.6, and Cav2.1α subtypes in the neurons of iron-induced epileptic rats.
Collapse
Affiliation(s)
- Chandra Prakash
- Neurobiology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Monika Mishra
- Neurobiology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Pavan Kumar
- Department of Developmental Neurogenetics, Medical University of South Carolina, Charleston, SC, USA
| | - Vikas Kumar
- Neurobiology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Deepak Sharma
- Neurobiology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
32
|
Young SM, Veeraraghavan P. Presynaptic voltage-gated calcium channels in the auditory brainstem. Mol Cell Neurosci 2021; 112:103609. [PMID: 33662542 DOI: 10.1016/j.mcn.2021.103609] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/06/2021] [Accepted: 02/17/2021] [Indexed: 10/22/2022] Open
Abstract
Sound information encoding within the initial synapses in the auditory brainstem requires reliable and precise synaptic transmission in response to rapid and large fluctuations in action potential (AP) firing rates. The magnitude and location of Ca2+ entry through voltage-gated Ca2+ channels (CaV) in the presynaptic terminal are key determinants in triggering AP-mediated release. In the mammalian central nervous system (CNS), the CaV2.1 subtype is the critical subtype for CNS function, since it is the most efficient CaV2 subtype in triggering AP-mediated synaptic vesicle (SV) release. Auditory brainstem synapses utilize CaV2.1 to sustain fast and repetitive SV release to encode sound information. Therefore, understanding the presynaptic mechanisms that control CaV2.1 localization, organization and biophysical properties are integral to understanding auditory processing. Here, we review our current knowledge about the control of presynaptic CaV2 abundance and organization in the auditory brainstem and impact on the regulation of auditory processing.
Collapse
Affiliation(s)
- Samuel M Young
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Department of Otolaryngology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA; Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| | | |
Collapse
|
33
|
Frankel EB, Kurshan PT. All Talk, No Assembly: Voltage-Gated Calcium Channels Do Not Mediate Active Zone Formation. Neuron 2020; 107:593-594. [PMID: 32818471 DOI: 10.1016/j.neuron.2020.07.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
How synapses assemble remains unknown. In this issue of Neuron, Held et al. (2020) demonstrate that Cav2-type voltage-gated calcium channels do not mediate presynaptic assembly. Moreover, the channel-associated protein α2δ localizes independently, suggesting additional functions for this auxiliary protein.
Collapse
Affiliation(s)
- Elisa B Frankel
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Peri T Kurshan
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
34
|
Meyer JO, Dahimene S, Page KM, Ferron L, Kadurin I, Ellaway JIJ, Zhao P, Patel T, Rothwell SW, Lin P, Pratt WS, Dolphin AC. Disruption of the Key Ca 2+ Binding Site in the Selectivity Filter of Neuronal Voltage-Gated Calcium Channels Inhibits Channel Trafficking. Cell Rep 2020; 29:22-33.e5. [PMID: 31577951 PMCID: PMC6899504 DOI: 10.1016/j.celrep.2019.08.079] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 06/20/2019] [Accepted: 08/22/2019] [Indexed: 12/05/2022] Open
Abstract
Voltage-gated calcium channels are exquisitely Ca2+ selective, conferred primarily by four conserved pore-loop glutamate residues contributing to the selectivity filter. There has been little previous work directly measuring whether the trafficking of calcium channels requires their ability to bind Ca2+ in the selectivity filter or to conduct Ca2+. Here, we examine trafficking of neuronal CaV2.1 and 2.2 channels with mutations in their selectivity filter and find reduced trafficking to the cell surface in cell lines. Furthermore, in hippocampal neurons, there is reduced trafficking to the somatic plasma membrane, into neurites, and to presynaptic terminals. However, the CaV2.2 selectivity filter mutants are still influenced by auxiliary α2δ subunits and, albeit to a reduced extent, by β subunits, indicating the channels are not grossly misfolded. Our results indicate that Ca2+ binding in the pore of CaV2 channels may promote their correct trafficking, in combination with auxiliary subunits. Furthermore, physiological studies utilizing selectivity filter mutant CaV channels should be interpreted with caution. Selectivity filter mutations in CaV2 channels block inward Ba2+ currents Surprisingly, these mutations severely reduce trafficking of the CaV2 channels Pore mutant N-type channels show reduced expression in presynaptic terminals Pore mutant channels still require β and α2δ and thus are not grossly misfolded
Collapse
Affiliation(s)
- James O Meyer
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Shehrazade Dahimene
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Karen M Page
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Laurent Ferron
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Ivan Kadurin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Joseph I J Ellaway
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Pengxiang Zhao
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Tarun Patel
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Simon W Rothwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Peipeng Lin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Wendy S Pratt
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK.
| |
Collapse
|
35
|
Mechanisms and Functional Consequences of Presynaptic Homeostatic Plasticity at Auditory Nerve Synapses. J Neurosci 2020; 40:6896-6909. [PMID: 32747441 DOI: 10.1523/jneurosci.1175-19.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 07/24/2020] [Accepted: 07/28/2020] [Indexed: 01/21/2023] Open
Abstract
Multiple forms of homeostasis influence synaptic function under diverse activity conditions. Both presynaptic and postsynaptic forms of homeostasis are important, but their relative impact on fidelity is unknown. To address this issue, we studied auditory nerve synapses onto bushy cells in the cochlear nucleus of mice of both sexes. These synapses undergo bidirectional presynaptic and postsynaptic homeostatic changes with increased and decreased acoustic stimulation. We found that both young and mature synapses exhibit similar activity-dependent changes in short-term depression. Experiments using chelators and imaging both indicated that presynaptic Ca2+ influx decreased after noise exposure, and increased after ligating the ear canal. By contrast, Ca2+ cooperativity was unaffected. Experiments using specific antagonists suggest that occlusion leads to changes in the Ca2+ channel subtypes driving neurotransmitter release. Furthermore, dynamic-clamp experiments revealed that spike fidelity primarily depended on changes in presynaptic depression, with some contribution from changes in postsynaptic intrinsic properties. These experiments indicate that presynaptic Ca2+ influx is homeostatically regulated in vivo to enhance synaptic fidelity.SIGNIFICANCE STATEMENT Homeostatic mechanisms in synapses maintain stable function in the face of different levels of activity. Both juvenile and mature auditory nerve synapses onto bushy cells modify short-term depression in different acoustic environments, which raises the question of what the underlying presynaptic mechanisms are and the relative importance of presynaptic and postsynaptic contributions to the faithful transfer of information. Changes in short-term depression under different acoustic conditions were a result of changes in presynaptic Ca2+ influx. Spike fidelity was affected by both presynaptic and postsynaptic changes after ear occlusion and was only affected by presynaptic changes after noise-rearing. These findings are important for understanding regulation of auditory synapses under normal conditions and also in disorders following noise exposure or conductive hearing loss.
Collapse
|
36
|
Held RG, Liu C, Ma K, Ramsey AM, Tarr TB, De Nola G, Wang SSH, Wang J, van den Maagdenberg AMJM, Schneider T, Sun J, Blanpied TA, Kaeser PS. Synapse and Active Zone Assembly in the Absence of Presynaptic Ca 2+ Channels and Ca 2+ Entry. Neuron 2020; 107:667-683.e9. [PMID: 32616470 DOI: 10.1016/j.neuron.2020.05.032] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 04/24/2020] [Accepted: 05/21/2020] [Indexed: 12/12/2022]
Abstract
Presynaptic CaV2 channels are essential for Ca2+-triggered exocytosis. In addition, there are two competing models for their roles in synapse structure. First, Ca2+ channels or Ca2+ entry may control synapse assembly. Second, active zone proteins may scaffold CaV2s to presynaptic release sites, and synapse structure is CaV2 independent. Here, we ablated all three CaV2s using conditional knockout in cultured hippocampal neurons or at the calyx of Held, which abolished evoked exocytosis. Compellingly, synapse and active zone structure, vesicle docking, and transsynaptic nano-organization were unimpaired. Similarly, long-term blockade of action potentials and Ca2+ entry did not disrupt active zone assembly. Although CaV2 knockout impaired the localization of β subunits, α2δ-1 localized normally. Rescue with CaV2 restored exocytosis, and CaV2 active zone targeting depended on the intracellular C-terminus. We conclude that synapse assembly is independent of CaV2s or Ca2+ entry through them. Instead, active zone proteins recruit and anchor CaV2s via CaV2 C-termini.
Collapse
Affiliation(s)
- Richard G Held
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Changliang Liu
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Kunpeng Ma
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA; State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, and University of Chinese Academy of Sciences, Beijing 100101, China
| | - Austin M Ramsey
- Department of Physiology and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Tyler B Tarr
- Department of Physiology and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Giovanni De Nola
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Shan Shan H Wang
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Jiexin Wang
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Toni Schneider
- Institute for Neurophysiology, University of Cologne, Köln 50931, Germany
| | - Jianyuan Sun
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, and University of Chinese Academy of Sciences, Beijing 100101, China
| | - Thomas A Blanpied
- Department of Physiology and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Pascal S Kaeser
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
37
|
Risher WC, Eroglu C. Emerging roles for α2δ subunits in calcium channel function and synaptic connectivity. Curr Opin Neurobiol 2020; 63:162-169. [PMID: 32521436 DOI: 10.1016/j.conb.2020.04.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/29/2020] [Indexed: 12/17/2022]
Abstract
Central nervous system function requires the proper formation and function of synapses. The α2δ auxiliary subunits of voltage-gated calcium channels have emerged as regulators of a number of critical events associated with regulation of synaptic function, including channel trafficking and localization, as well as the initial establishment of synaptic structures. In this review, we will discuss some of these recent studies which have uncovered novel mechanisms for α2δ function at the synapse, including the regulation of calcium channel α1 subunit specificity and the promotion of dendritic spine growth. Moreover, we will cover recent advances that have been made in understanding the consequences of aberrant α2δ signaling in injury and disease.
Collapse
Affiliation(s)
- William Christopher Risher
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine at Marshall University, Huntington, WV 25705, United States.
| | - Cagla Eroglu
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, United States; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, United States; Duke Institute for Brain Sciences (DIBS), Durham, NC 27710, United States; Regeneration Next Initiative, Duke University, Durham, NC 27710, United States
| |
Collapse
|
38
|
Wang S, Cortes CJ. Interactions with PDZ proteins diversify voltage-gated calcium channel signaling. J Neurosci Res 2020; 99:332-348. [PMID: 32476168 DOI: 10.1002/jnr.24650] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 04/29/2020] [Accepted: 05/07/2020] [Indexed: 11/12/2022]
Abstract
Voltage-gated Ca2+ (CaV ) channels are crucial for neuronal excitability and synaptic transmission upon depolarization. Their properties in vivo are modulated by their interaction with a variety of scaffolding proteins. Such interactions can influence the function and localization of CaV channels, as well as their coupling to intracellular second messengers and regulatory pathways, thus amplifying their signaling potential. Among these scaffolding proteins, a subset of PDZ (postsynaptic density-95, Drosophila discs-large, and zona occludens)-domain containing proteins play diverse roles in modulating CaV channel properties. At the presynaptic terminal, PDZ proteins enrich CaV channels in the active zone, enabling neurotransmitter release by maintaining a tight and vital link between channels and vesicles. In the postsynaptic density, these interactions are essential in regulating dendritic spine morphology and postsynaptic signaling cascades. In this review, we highlight the studies that demonstrate dynamic regulations of neuronal CaV channels by PDZ proteins. We discuss the role of PDZ proteins in controlling channel activity, regulating channel cell surface density, and influencing channel-mediated downstream signaling events. We highlight the importance of PDZ protein regulations of CaV channels and evaluate the link between this regulatory effect and human disease.
Collapse
Affiliation(s)
- Shiyi Wang
- Department of Cell Biology, Duke University, Durham, NC, USA.,Department of Neurology, Duke University, Durham, NC, USA
| | - Constanza J Cortes
- Department of Neurology, Duke University, Durham, NC, USA.,Department of Cell, Developmental and Integrative Biology, University of Alabama Birmingham, Birmingham, AL, USA
| |
Collapse
|
39
|
Radulovic T, Dong W, Goral RO, Thomas CI, Veeraraghavan P, Montesinos MS, Guerrero-Given D, Goff K, Lübbert M, Kamasawa N, Ohtsuka T, Young SM. Presynaptic development is controlled by the core active zone proteins CAST/ELKS. J Physiol 2020; 598:2431-2452. [PMID: 32304329 DOI: 10.1113/jp279736] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 04/14/2020] [Indexed: 12/24/2022] Open
Abstract
KEY POINTS CAST/ELKS are positive regulators of presynaptic growth and are suppressors of active zone expansion at the developing mouse calyx of Held. CAST/ELKS regulate all three CaV 2 subtype channel levels in the presynaptic terminal and not just CaV 2.1. The half-life of ELKS is on the timescale of days and not weeks. Synaptic transmission was not impacted by the loss of CAST/ELKS. CAST/ELKS are involved in pathways regulating morphological properties of presynaptic terminals during an early stage of circuit maturation. ABSTRACT Many presynaptic active zone (AZ) proteins have multiple regulatory roles that vary during distinct stages of neuronal circuit development. The CAST/ELKS protein family are evolutionarily conserved presynaptic AZ molecules that regulate presynaptic calcium channels, synaptic transmission and plasticity in the mammalian CNS. However, how these proteins regulate synapse development and presynaptic function in a developing neuronal circuit in its native environment is unclear. To unravel the roles of CAST/ELKS in glutamatergic synapse development and in presynaptic function, we used CAST knockout (KO) and ELKS conditional KO (CKO) mice to examine how their loss during the early stages of circuit maturation impacted the calyx of Held presynaptic terminal development and function. Morphological analysis from confocal z-stacks revealed that combined deletion of CAST/ELKS resulted in a reduction in the surface area and volume of the calyx. Analysis of AZ ultrastructure showed that AZ size was increased in the absence of CAST/ELKS. Patch clamp recordings demonstrated a reduction of all presynaptic CaV 2 channel subtype currents that correlated with a loss in presynaptic CaV 2 channel numbers. However, these changes did not impair synaptic transmission and plasticity and synaptic vesicle release kinetics. We conclude that CAST/ELKS proteins are positive regulators of presynaptic growth and are suppressors of AZ expansion and CaV 2 subtype currents and levels during calyx of Held development. We propose that CAST/ELKS are involved in pathways regulating presynaptic morphological properties and CaV 2 channel subtypes and suggest there is developmental compensation to preserve synaptic transmission during early stages of neuronal circuit maturation.
Collapse
Affiliation(s)
- Tamara Radulovic
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Wei Dong
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Medical Electrophysiological Key Lab of Sichuan Province, Institute of Cardiovascular Research of Southwest Medical University, Luzhou, 646000, China
| | - R Oliver Goral
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Connon I Thomas
- Electron Microscopy Facility, Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
| | | | - Monica Suarez Montesinos
- Research Group Molecular Mechanisms of Synaptic Function, Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
| | - Debbie Guerrero-Given
- Electron Microscopy Facility, Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
| | - Kevin Goff
- Research Group Molecular Mechanisms of Synaptic Function, Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
| | - Matthias Lübbert
- Research Group Molecular Mechanisms of Synaptic Function, Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
| | - Naomi Kamasawa
- Electron Microscopy Facility, Max Planck Florida Institute for Neuroscience, Jupiter, FL, 33458, USA
| | - Toshihisa Ohtsuka
- Department of Biochemistry , Graduate School of Medicine/Faculty of Medicine, University of Yamanashi, 1110 Shimokato Chuo, Yamanashi, 409-3898, Japan
| | - Samuel M Young
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA.,Department of Otolaryngology, Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| |
Collapse
|
40
|
Assembly of the presynaptic active zone. Curr Opin Neurobiol 2020; 63:95-103. [PMID: 32403081 DOI: 10.1016/j.conb.2020.03.008] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/29/2020] [Accepted: 03/18/2020] [Indexed: 12/11/2022]
Abstract
In a presynaptic nerve terminal, the active zone is composed of sophisticated protein machinery that enables secretion on a submillisecond time scale and precisely targets it toward postsynaptic receptors. The past two decades have provided deep insight into the roles of active zone proteins in exocytosis, but we are only beginning to understand how a neuron assembles active zone protein complexes into effective molecular machines. In this review, we outline the fundamental processes that are necessary for active zone assembly and discuss recent advances in understanding assembly mechanisms that arise from genetic, morphological and biochemical studies. We further outline the challenges ahead for understanding this important problem.
Collapse
|
41
|
Xiao Y, Luo H, Yang WZ, Zeng Y, Shen Y, Ni X, Shi Z, Zhong J, Liang Z, Fu X, Tu H, Sun W, Shen WL, Hu J, Yang J. A Brain Signaling Framework for Stress-Induced Depression and Ketamine Treatment Elucidated by Phosphoproteomics. Front Cell Neurosci 2020; 14:48. [PMID: 32317933 PMCID: PMC7156020 DOI: 10.3389/fncel.2020.00048] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 02/20/2020] [Indexed: 12/25/2022] Open
Abstract
Depression is a common affective disorder characterized by significant and persistent low mood. Ketamine, an N-methyl-D-aspartate receptor (NMDAR) antagonist, is reported to have a rapid and durable antidepressant effect, but the mechanisms are unclear. Protein phosphorylation is a post-translational modification that plays a crucial role in cell signaling. Thus, we present a phosphoproteomics approach to investigate the mechanisms underlying stress-induced depression and the rapid antidepressant effect of ketamine in mice. We analyzed the phosphoprotein changes induced by chronic unpredictable mild stress (CUMS) and ketamine treatment in two known mood control centers, the medial prefrontal cortex (mPFC) and the nucleus accumbens (NAc). We initially obtained >8,000 phosphorylation sites. Quantitation revealed 3,988 sites from the mPFC and 3,196 sites from the NAc. Further analysis revealed that changes in synaptic transmission-related signaling are a common feature. Notably, CUMS-induced changes were reversed by ketamine treatment, as shown by the analysis of commonly altered sites. Ketamine also induced specific changes, such as alterations in synapse organization, synaptic transmission, and enzyme binding. Collectively, our findings establish a signaling framework for stress-induced depression and the rapid antidepressant effect of ketamine.
Collapse
Affiliation(s)
- Yan Xiao
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Huoqing Luo
- School of Life Science and Technology, Shanghaitech University, Shanghai, China.,State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Institute of Neuroscience, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wen Z Yang
- Shanghai Institute for Advanced Immunochemical Studies & School of Life Science and Technology, Shanghaitech University, Shanghai, China.,CAS Key Laboratory of Synthetic Chemistry of Natural Substances, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Yeting Zeng
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yinbo Shen
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xinyan Ni
- School of Life Science and Technology, Shanghaitech University, Shanghai, China
| | - Zhaomei Shi
- School of Life Science and Technology, Shanghaitech University, Shanghai, China
| | - Jun Zhong
- Delta Omics Inc., Baltimore, MD, United States
| | - Ziqi Liang
- School of Life Science and Technology, Shanghaitech University, Shanghai, China
| | - Xiaoyu Fu
- School of Life Science and Technology, Shanghaitech University, Shanghai, China.,State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Institute of Neuroscience, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Hongqing Tu
- School of Life Science and Technology, Shanghaitech University, Shanghai, China.,State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Institute of Neuroscience, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wenzhi Sun
- Chinese Institute For Brain Research, Beijing, China
| | - Wei L Shen
- School of Life Science and Technology, Shanghaitech University, Shanghai, China
| | - Ji Hu
- School of Life Science and Technology, Shanghaitech University, Shanghai, China
| | - Jiajun Yang
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
42
|
Dolphin AC, Lee A. Presynaptic calcium channels: specialized control of synaptic neurotransmitter release. Nat Rev Neurosci 2020; 21:213-229. [PMID: 32161339 PMCID: PMC7873717 DOI: 10.1038/s41583-020-0278-2] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2020] [Indexed: 11/09/2022]
Abstract
Chemical synapses are heterogeneous junctions formed between neurons that are specialized for the conversion of electrical impulses into the exocytotic release of neurotransmitters. Voltage-gated Ca2+ channels play a pivotal role in this process as they are the major conduits for the Ca2+ ions that trigger the fusion of neurotransmitter-containing vesicles with the presynaptic membrane. Alterations in the intrinsic function of these channels and their positioning within the active zone can profoundly alter the timing and strength of synaptic output. Advances in optical and electron microscopic imaging, structural biology and molecular techniques have facilitated recent breakthroughs in our understanding of the properties of voltage-gated Ca2+ channels that support their presynaptic functions. Here we examine the nature of these channels, how they are trafficked to and anchored within presynaptic boutons, and the mechanisms that allow them to function optimally in shaping the flow of information through neural circuits.
Collapse
Affiliation(s)
- Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK.
| | - Amy Lee
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
43
|
Ferron L, Novazzi CG, Pilch KS, Moreno C, Ramgoolam K, Dolphin AC. FMRP regulates presynaptic localization of neuronal voltage gated calcium channels. Neurobiol Dis 2020; 138:104779. [PMID: 31991246 PMCID: PMC7152798 DOI: 10.1016/j.nbd.2020.104779] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 01/09/2020] [Accepted: 01/24/2020] [Indexed: 12/31/2022] Open
Abstract
Fragile X syndrome (FXS), the most common form of inherited intellectual disability and autism, results from the loss of fragile X mental retardation protein (FMRP). We have recently identified a direct interaction of FMRP with voltage-gated Ca2+ channels that modulates neurotransmitter release. In the present study we used a combination of optophysiological tools to investigate the impact of FMRP on the targeting of voltage-gated Ca2+ channels to the active zones in neuronal presynaptic terminals. We monitored Ca2+ transients at synaptic boutons of dorsal root ganglion (DRG) neurons using the genetically-encoded Ca2+ indicator GCaMP6f tagged to synaptophysin. We show that knock-down of FMRP induces an increase of the amplitude of the Ca2+ transient in functionally-releasing presynaptic terminals, and that this effect is due to an increase of N-type Ca2+ channel contribution to the total Ca2+ transient. Dynamic regulation of CaV2.2 channel trafficking is key to the function of these channels in neurons. Using a CaV2.2 construct with an α-bungarotoxin binding site tag, we further investigate the impact of FMRP on the trafficking of CaV2.2 channels. We show that forward trafficking of CaV2.2 channels from the endoplasmic reticulum to the plasma membrane is reduced when co-expressed with FMRP. Altogether our data reveal a critical role of FMRP on localization of CaV channels to the presynaptic terminals and how its defect in a context of FXS can profoundly affect synaptic transmission. Loss of FMRP increases presynaptic Ca2+ transients. FMRP is a negative regulator of presynaptic Cav2.2 channel abundance. FMRP reduces the forward trafficking of Cav2.2 channels from ER to plasma membrane. Distal part of FMRP carboxy terminus is key for interaction with Cav2.2 channels.
Collapse
Affiliation(s)
- Laurent Ferron
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK.
| | - Cesare G Novazzi
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Kjara S Pilch
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Cristian Moreno
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Krishma Ramgoolam
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| |
Collapse
|
44
|
Quantitation and Simulation of Single Action Potential-Evoked Ca 2+ Signals in CA1 Pyramidal Neuron Presynaptic Terminals. eNeuro 2019; 6:ENEURO.0343-19.2019. [PMID: 31551250 PMCID: PMC6800293 DOI: 10.1523/eneuro.0343-19.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 09/10/2019] [Indexed: 01/07/2023] Open
Abstract
Presynaptic Ca2+ evokes exocytosis, endocytosis, and synaptic plasticity. However, Ca2+ flux and interactions at presynaptic molecular targets are difficult to quantify because fluorescence imaging has limited resolution. In rats of either sex, we measured single varicosity presynaptic Ca2+ using Ca2+ dyes as buffers, and constructed models of Ca2+ dispersal. Action potentials evoked Ca2+ transients with little variation when measured with low-affinity dye (peak amplitude 789 ± 39 nM, within 2 ms of stimulation; decay times, 119 ± 10 ms). Endogenous Ca2+ buffering capacity, action potential-evoked free [Ca2+]i, and total Ca2+ amounts entering terminals were determined using Ca2+ dyes as buffers. These data constrained Monte Carlo (MCell) simulations of Ca2+ entry, buffering, and removal. Simulations of experimentally-determined Ca2+ fluxes, buffered by simulated calbindin28K well fit data, and were consistent with clustered Ca2+ entry followed within 4 ms by diffusion throughout the varicosity. Repetitive stimulation caused free varicosity Ca2+ to sum. However, simulated in nanometer domains, its removal by pumps and buffering was negligible, while local diffusion dominated. Thus, Ca2+ within tens of nanometers of entry, did not accumulate. A model of synaptotagmin1 (syt1)-Ca2+ binding indicates that even with 10 µM free varicosity evoked Ca2+, syt1 must be within tens of nanometers of channels to ensure occupation of all its Ca2+-binding sites. Repetitive stimulation, evoking short-term synaptic enhancement, does not modify probabilities of Ca2+ fully occupying syt1’s C2 domains, suggesting that enhancement is not mediated by Ca2+-syt1 interactions. We conclude that at spatiotemporal scales of fusion machines, Ca2+ necessary for their activation is diffusion dominated.
Collapse
|
45
|
Stephani F, Scheuer V, Eckrich T, Blum K, Wang W, Obermair GJ, Engel J. Deletion of the Ca 2+ Channel Subunit α 2δ3 Differentially Affects Ca v2.1 and Ca v2.2 Currents in Cultured Spiral Ganglion Neurons Before and After the Onset of Hearing. Front Cell Neurosci 2019; 13:278. [PMID: 31293392 PMCID: PMC6606706 DOI: 10.3389/fncel.2019.00278] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 06/07/2019] [Indexed: 12/14/2022] Open
Abstract
Voltage-gated Ca2+ channels are composed of a pore-forming α1 subunit and auxiliary β and α2δ subunits, which modulate Ca2+ current properties and channel trafficking. So far, the partial redundancy and specificity of α1 for α2δ subunits in the CNS have remained largely elusive. Mature spiral ganglion (SG) neurons express α2δ subunit isoforms 1, 2, and 3 and multiple Ca2+ channel subtypes. Differentiation and in vivo functions of their endbulb of Held synapses, which rely on presynaptic P/Q channels (Lin et al., 2011), require the α2δ3 subunit (Pirone et al., 2014). This led us to hypothesize that P/Q channels may preferentially co-assemble with α2δ3. Using a dissociated primary culture, we analyzed the effects of α2δ3 deletion on somatic Ca2+ currents (ICa) of SG neurons isolated at postnatal day 20 (P20), when the cochlea is regarded to be mature. P/Q currents were the dominating steady-state Ca2+ currents (54% of total) followed by T-type, L-type, N-type, and R-type currents. Deletion of α2δ3 reduced P/Q- and R-type currents by 60 and 38%, respectively, whereas L-type, N-type, and T-type currents were not altered. A subset of ICa types was also analyzed in SG neurons isolated at P5, i.e., before the onset of hearing (P12). Both L-type and N-type current amplitudes of wildtype SG neurons were larger at P5 compared with P20. Deletion of α2δ3 reduced L-type and N-type currents by 23 and 44%, respectively. In contrast, small P/Q currents, which were just being up-regulated at P5, were unaffected by the lack of α2δ3. In summary, α2δ3 regulates amplitudes of L- and N-type currents of immature cultured SG neurons, whereas it regulates P/Q- and R-type currents at P20. Our data indicate a developmental switch from dominating somatic N- to P/Q-type currents in cultured SG neurons. A switch from N- to P/Q-type channels, which has been observed at several central synapses, may also occur at developing endbulbs of Held. In this case, reduction of both neonatal N- (P5) and more mature P/Q-type currents (around/after hearing onset) may contribute to the impaired morphology and function of endbulb synapses in α2δ3-deficient mice.
Collapse
Affiliation(s)
- Friederike Stephani
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, Homburg, Germany
| | - Veronika Scheuer
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, Homburg, Germany
| | - Tobias Eckrich
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, Homburg, Germany
| | - Kerstin Blum
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, Homburg, Germany
| | - Wenying Wang
- Department of Physiology, School of Medicine, University of Nevada, Reno, NV, United States
| | - Gerald J Obermair
- Department of Physiology and Medical Physics, Medical University Innsbruck, Innsbruck, Austria.,Division Physiology, Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Jutta Engel
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, Homburg, Germany
| |
Collapse
|
46
|
Tyagi S, Bendrick TR, Filipova D, Papadopoulos S, Bannister RA. A mutation in Ca V2.1 linked to a severe neurodevelopmental disorder impairs channel gating. J Gen Physiol 2019; 151:850-859. [PMID: 31015257 PMCID: PMC6571999 DOI: 10.1085/jgp.201812237] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 02/04/2019] [Accepted: 03/18/2019] [Indexed: 01/07/2023] Open
Abstract
Ca2+ flux into axon terminals via P-/Q-type CaV2.1 channels is the trigger for neurotransmitter vesicle release at neuromuscular junctions (NMJs) and many central synapses. Recently, an arginine to proline substitution (R1673P) in the S4 voltage-sensing helix of the fourth membrane-bound repeat of CaV2.1 was linked to a severe neurological disorder characterized by generalized hypotonia, ataxia, cerebellar atrophy, and global developmental delay. The R1673P mutation was proposed to cause a gain of function in CaV2.1 leading to neuronal Ca2+ toxicity based on the ability of the mutant channel to rescue the photoreceptor response in CaV2.1-deficient Drosophila cacophony larvae. Here, we show that the corresponding mutation in rat CaV2.1 (R1624P) causes a profound loss of channel function; voltage-clamp analysis of tsA-201 cells expressing this mutant channel revealed an ∼25-mV depolarizing shift in the voltage dependence of activation. This alteration in activation implies that a significant fraction of CaV2.1 channels resident in presynaptic terminals are unlikely to open in response to an action potential, thereby increasing the probability of synaptic failure at both NMJs and central synapses. Indeed, the mutant channel supported only minimal Ca2+ flux in response to an action potential-like waveform. Application of GV-58, a compound previously shown to stabilize the open state of wild-type CaV2.1 channels, partially restored Ca2+ current by shifting mutant activation to more hyperpolarizing potentials and slowing deactivation. Consequently, GV-58 also rescued a portion of Ca2+ flux during action potential-like stimuli. Thus, our data raise the possibility that therapeutic agents that increase channel open probability or prolong action potential duration may be effective in combatting this and other severe neurodevelopmental disorders caused by loss-of-function mutations in CaV2.1.
Collapse
Affiliation(s)
- Sidharth Tyagi
- Department of Medicine-Cardiology Division, University of Colorado School of Medicine, Aurora, CO
| | - Tyler R Bendrick
- Department of Medicine-Cardiology Division, University of Colorado School of Medicine, Aurora, CO
| | - Dilyana Filipova
- Department of Vegetative Physiology, University of Cologne, Cologne, Germany
| | - Symeon Papadopoulos
- Department of Vegetative Physiology, University of Cologne, Cologne, Germany
| | - Roger A Bannister
- Department of Medicine-Cardiology Division, University of Colorado School of Medicine, Aurora, CO
| |
Collapse
|
47
|
Goel P, Dufour Bergeron D, Böhme MA, Nunnelly L, Lehmann M, Buser C, Walter AM, Sigrist SJ, Dickman D. Homeostatic scaling of active zone scaffolds maintains global synaptic strength. J Cell Biol 2019; 218:1706-1724. [PMID: 30914419 PMCID: PMC6504899 DOI: 10.1083/jcb.201807165] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 12/14/2018] [Accepted: 03/06/2019] [Indexed: 12/23/2022] Open
Abstract
Synaptic terminals grow and retract throughout life, yet synaptic strength is maintained within stable physiological ranges. To study this process, we investigated Drosophila endophilin (endo) mutants. Although active zone (AZ) number is doubled in endo mutants, a compensatory reduction in their size homeostatically adjusts global neurotransmitter output to maintain synaptic strength. We find an inverse adaptation in rab3 mutants. Additional analyses using confocal, STED, and electron microscopy reveal a stoichiometric tuning of AZ scaffolds and nanoarchitecture. Axonal transport of synaptic cargo via the lysosomal kinesin adapter Arl8 regulates AZ abundance to modulate global synaptic output and sustain the homeostatic potentiation of neurotransmission. Finally, we find that this AZ scaling can interface with two independent homeostats, depression and potentiation, to remodel AZ structure and function, demonstrating a robust balancing of separate homeostatic adaptations. Thus, AZs are pliable substrates with elastic and modular nanostructures that can be dynamically sculpted to stabilize and tune both local and global synaptic strength.
Collapse
Affiliation(s)
- Pragya Goel
- Department of Neurobiology, University of Southern California, Los Angeles, CA
| | | | - Mathias A Böhme
- Neurocure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
| | - Luke Nunnelly
- Department of Neurobiology, University of Southern California, Los Angeles, CA
| | - Martin Lehmann
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | | | - Alexander M Walter
- Neurocure Cluster of Excellence, Charité Universitätsmedizin, Berlin, Germany
| | | | - Dion Dickman
- Department of Neurobiology, University of Southern California, Los Angeles, CA
| |
Collapse
|