1
|
Dar NJ, Currais A, Taguchi T, Andrews N, Maher P. Cannabinol (CBN) alleviates age-related cognitive decline by improving synaptic and mitochondrial health. Redox Biol 2025; 84:103692. [PMID: 40412024 DOI: 10.1016/j.redox.2025.103692] [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: 02/21/2025] [Revised: 05/16/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025] Open
Abstract
Age-related cognitive decline and neurodegenerative diseases, such as Alzheimer's disease, represent major global health challenges, particularly with an aging population. Mitochondrial dysfunction appears to play a central role in the pathophysiology of these conditions by driving redox dysregulation and impairing cellular energy metabolism. Despite extensive research, effective therapeutic options remain limited. Cannabinol (CBN), a cannabinoid previously identified as a potent inhibitor of oxytosis/ferroptosis through mitochondrial modulation, has demonstrated promising neuroprotective effects. In cell culture, CBN targets mitochondria, preserving mitochondrial membrane potential, enhancing antioxidant defenses and regulating bioenergetic processes. However, the in vivo therapeutic potential of CBN, particularly in aging models, has not been thoroughly explored. To address this gap, this study investigated the effects of CBN on age-associated cognitive decline and metabolic dysfunction using the SAMP8 mouse model of accelerated aging. Our results show that CBN significantly improves spatial learning and memory, with more pronounced cognitive benefits observed in female mice. These cognitive improvements are accompanied by sex-specific changes in metabolic parameters, such as enhanced oxygen consumption and energy expenditure. Mechanistically, CBN modulates key regulators of mitochondrial dynamics, including mitofusin 2 (MFN2) and dynamin-related protein 1 (DRP1), while upregulating markers of mitochondrial biogenesis including mitochondrial transcription factor A (TFAM) and translocase of outer mitochondrial membrane 20 (TOM20). Additionally, CBN upregulates key synaptic proteins involved in vesicle trafficking and postsynaptic signaling suggesting that it enhances synaptic function and neurotransmission, further reinforcing its neuroprotective effects. This study provides in vivo evidence supporting CBN's potential to mitigate age-related cognitive and metabolic dysfunction, with notable sex-specific effects, highlighting its promise for neurodegenerative diseases and cognitive decline.
Collapse
Affiliation(s)
- Nawab John Dar
- Department of Cellular Neurobiology, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Antonio Currais
- Department of Cellular Neurobiology, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Taketo Taguchi
- Department of Cellular Neurobiology, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Nick Andrews
- Behaviour Testing Core, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Pamela Maher
- Department of Cellular Neurobiology, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
2
|
Pang Y, Wang C, Zhang YZ, Wang Z, Imoto S, Lee TY. STForte: tissue context-specific encoding and consistency-aware spatial imputation for spatially resolved transcriptomics. Brief Bioinform 2025; 26:bbaf174. [PMID: 40254832 PMCID: PMC12009714 DOI: 10.1093/bib/bbaf174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/06/2025] [Accepted: 03/17/2025] [Indexed: 04/22/2025] Open
Abstract
Encoding spatially resolved transcriptomics (SRT) data serves to identify the biological semantics of RNA expression within the tissue while preserving spatial characteristics. Depending on the analytical scenario, one may focus on different contextual structures of tissues. For instance, anatomical regions reveal consistent patterns by focusing on spatial homogeneity, while elucidating complex tumor micro-environments requires more expression heterogeneity. However, current spatial encoding methods lack consideration of the tissue context. Meanwhile, most developed SRT technologies are still limited in providing exact patterns of intact tissues due to limitations such as low resolution or missed measurements. Here, we propose STForte, a novel pairwise graph autoencoder-based approach with cross-reconstruction and adversarial distribution matching, to model the spatial homogeneity and expression heterogeneity of SRT data. STForte extracts interpretable latent encodings, enabling downstream analysis by accurately portraying various tissue contexts. Moreover, STForte allows spatial imputation using only spatial consistency to restore the biological patterns of unobserved locations or low-quality cells, thereby providing fine-grained views to enhance the SRT analysis. Extensive evaluations of datasets under different scenarios and SRT platforms demonstrate that STForte is a scalable and versatile tool for providing enhanced insights into spatial data analysis.
Collapse
Affiliation(s)
- Yuxuan Pang
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Chunxuan Wang
- School of Data Science, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), 2001 Longxiang Road, Longgang, Shenzhen, 518172, China
| | - Yao-zhong Zhang
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Zhuo Wang
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), 2001 Longxiang Road, Longgang, Shenzhen, 518172, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), 2001 Longxiang Road, Longgang, Shenzhen, 518172, China
| | - Seiya Imoto
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Tzong-Yi Lee
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, No. 75 Bo-Ai Street, Hsinchu 300, Taiwan
| |
Collapse
|
3
|
Hayoz S, Jia C, Hegg CC. Constitutive and evoked release of ATP in adult mouse olfactory epithelium. Open Life Sci 2024; 19:20220811. [PMID: 38250473 PMCID: PMC10795008 DOI: 10.1515/biol-2022-0811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/31/2023] [Accepted: 11/28/2023] [Indexed: 01/23/2024] Open
Abstract
In adult olfactory epithelium (OE), ATP plays a role in constant cell turnover and post-injury neuroregeneration. We previously demonstrated that constitutive and ATP-evoked ATP release are present in neonatal mouse OE and underlie continuous cell turn-over and post-injury neuroregeneration, and that activation of purinergic P2X7 receptors is involved in the evoked release. We hypothesized that both releases are present in adult mouse OE. To study the putative contribution of olfactory sensory neurons to ATP release, we used olfactory sensory neuronal-like OP6 cells derived from the embryonic olfactory placode cells. Calcium imaging showed that OP6 cells and primary adult OE cell cultures express functional purinergic receptors. We monitored ATP release from OP6 cells and whole adult OE turbinates using HEK cells as biosensors and luciferin-luciferase assays. Constitutive ATP release occurs in OP6 cells and whole adult mouse OE turbinates, and P2X7 receptors mediated evoked ATP release occurs only in turbinates. The mechanisms of ATP release described in the present study might underlie the constant cell turn-over and post-injury neuroregeneration present in adult OE and thus, further studies of these mechanisms are warranted as it will improve our knowledge of OE tissue homeostasis and post-injury regeneration.
Collapse
Affiliation(s)
- Sébastien Hayoz
- Department of Physiology, University of Arizona, Tucson, Arizona 85724, USA
| | - Cuihong Jia
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee 37614, USA
| | - Colleen Cosgrove Hegg
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan 48824, USA
| |
Collapse
|
4
|
Kao Y, Zhu H, Yang Y, Shen W, Song W, Zhang R, Liu Y, Liu H, Kong X. CREB1 Facilitates GABAergic Neural Differentiation of Human Mesenchymal Stem Cells through BRN2 for Pain Alleviation and Locomotion Recovery after Spinal Cord Injury. Cells 2023; 13:67. [PMID: 38201271 PMCID: PMC10778540 DOI: 10.3390/cells13010067] [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: 09/30/2023] [Revised: 11/28/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
The transplantation of GABAergic neuron cells has been reported to alleviate nerve pain and improve motor function after spinal cord injury (SCI). However, human mesenchymal stem cell (hMSC) differentiation into GABAergic neuron cells in a sufficient quantity remains to be accomplished. From a database screening, cAMP-responsive element-binding protein 1 (CREB1) was chosen as a potential modulator due to its critical role in the protein-protein interaction of genes related to GABAergic neural differentiation. Here, CREB1 was overexpressed in transfected hMSCs, where CREB1 could induce differentiation into GABAergic neuron cells with an upregulation of Map2 and GAD1 by 2- and 3.4-fold, respectively. Additionally, GABAergic neural differentiation was enhanced, while Notch signaling was inhibited, and BRN2 transcriptional activation played an important role in neuronal maturation. Moreover, transfected hMSCs injected into immunocompromised mice caused by CsA exhibited the neuronal markers Tuj1 and Map2 via the intraspinal route, suggesting an improvement in survival and neural differentiation. Significantly, improvement in both BMS scores (6.2 ± 1.30 vs. 4 ± 0) and thermal hyperalgesia latency (7.74 ± 2.36 s vs. 4.52 ± 0.39 s) was seen compared with the SCI naïve treatment at 4 weeks post-transplantation. Our study demonstrates that CREB1 is crucial in generating induced GABAergic neuron cells (iGNs) originating from hMSCs. Transplanting iGNs to injured spinal cord provides a promising strategy for alleviating neuropathic pain and locomotion recovery after SCI.
Collapse
Affiliation(s)
- Yanbing Kao
- Orthopedic Research Center of Qilu Hospital, Shandong University, Jinan 250100, China
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
| | - Hanming Zhu
- Orthopedic Research Center of Qilu Hospital, Shandong University, Jinan 250100, China
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
| | - Yu Yang
- Orthopedic Research Center of Qilu Hospital, Shandong University, Jinan 250100, China
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
| | - Wenyuan Shen
- Orthopedic Research Center of Qilu Hospital, Shandong University, Jinan 250100, China
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
| | - Wei Song
- Orthopedic Research Center of Qilu Hospital, Shandong University, Jinan 250100, China
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
| | - Renjie Zhang
- Orthopedic Research Center of Qilu Hospital, Shandong University, Jinan 250100, China
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
| | - Yanchun Liu
- Orthopedic Research Center of Qilu Hospital, Shandong University, Jinan 250100, China
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
| | - Haoyun Liu
- Orthopedic Research Center of Qilu Hospital, Shandong University, Jinan 250100, China
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
| | - Xiaohong Kong
- Orthopedic Research Center of Qilu Hospital, Shandong University, Jinan 250100, China
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan 250100, China
| |
Collapse
|
5
|
Chen D, Zhang Y, Qiao R, Kong X, Zhong H, Wang X, Zhu J, Li B. Integrated bioinformatics-based identification of diagnostic markers in Alzheimer disease. Front Aging Neurosci 2022; 14:988143. [PMID: 36437991 PMCID: PMC9686423 DOI: 10.3389/fnagi.2022.988143] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 10/28/2022] [Indexed: 08/09/2023] Open
Abstract
Alzheimer disease (AD) is a progressive neurodegenerative disease resulting from the accumulation of extracellular amyloid beta (Aβ) and intracellular neurofibrillary tangles. There are currently no objective diagnostic measures for AD. The aim of this study was to identify potential diagnostic markers for AD and evaluate the role of immune cell infiltration in disease pathogenesis. AD expression profiling data for human hippocampus tissue (GSE48350 and GSE5281) were downloaded from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) were identified using R software and the Human Protein Atlas database was used to screen AD-related DEGs. We performed functional enrichment analysis and established a protein-protein interaction (PPI) network to identify disease-related hub DEGs. The fraction of infiltrating immune cells in samples was determined with the Microenvironment Cell Populations-counter method. The random forest algorithm was used to develop a prediction model and receiver operating characteristic (ROC) curve analysis was performed to validate the diagnostic utility of the candidate AD markers. The correlation between expression of the diagnostic markers and immune cell infiltration was also analyzed. A total of 107 AD-related DEGs were screened in this study, including 28 that were upregulated and 79 that were downregulated. The DEGs were enriched in the Gene Ontology terms GABAergic synapse, Morphine addiction, Nicotine addiction, Phagosome, and Synaptic vesicle cycle. We identified 10 disease-related hub genes and 20 candidate diagnostic genes. Synaptophysin (SYP) and regulator of G protein signaling 4 (RGS4) (area under the ROC curve = 0.909) were verified as potential diagnostic markers for AD in the GSE28146 validation dataset. Natural killer cells, B lineage cells, monocytic lineage cells, endothelial cells, and fibroblasts were found to be involved in AD; additionally, the expression levels of both SYP and RGS4 were negatively correlated with the infiltration of these immune cell types. These results suggest that SYP and RGS4 are potential diagnostic markers for AD and that immune cell infiltration plays an important role in AD development and progression.
Collapse
Affiliation(s)
- Danmei Chen
- Research Center for Clinical Medicine, Jinshan Hospital Affiliated to Fudan University, Shanghai, China
- Department of Integrative Medicine, Huashan Hospital Affiliated to Fudan University, Shanghai, China
| | - Yunpeng Zhang
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Rui Qiao
- College of Acupuncture-Massage and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Xiangyu Kong
- Research Center for Clinical Medicine, Jinshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Hequan Zhong
- Research Center for Clinical Medicine, Jinshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Xiaokun Wang
- Research Center for Clinical Medicine, Jinshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Jie Zhu
- Department of Rehabilitation, Jinshan Hospital, Fudan University, Shanghai, China
| | - Bing Li
- Research Center for Clinical Medicine, Jinshan Hospital Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
6
|
Phongpreecha T, Gajera CR, Liu CC, Vijayaragavan K, Chang AL, Becker M, Fallahzadeh R, Fernandez R, Postupna N, Sherfield E, Tebaykin D, Latimer C, Shively CA, Register TC, Craft S, Montine KS, Fox EJ, Poston KL, Keene CD, Angelo M, Bendall SC, Aghaeepour N, Montine TJ. Single-synapse analyses of Alzheimer's disease implicate pathologic tau, DJ1, CD47, and ApoE. SCIENCE ADVANCES 2021; 7:eabk0473. [PMID: 34910503 PMCID: PMC8673771 DOI: 10.1126/sciadv.abk0473] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Synaptic molecular characterization is limited for Alzheimer’s disease (AD). Our newly invented mass cytometry–based method, synaptometry by time of flight (SynTOF), was used to measure 38 antibody probes in approximately 17 million single-synapse events from human brains without pathologic change or with pure AD or Lewy body disease (LBD), nonhuman primates (NHPs), and PS/APP mice. Synaptic molecular integrity in humans and NHP was similar. Although not detected in human synapses, Aβ was in PS/APP mice single-synapse events. Clustering and pattern identification of human synapses showed expected disease-specific differences, like increased hippocampal pathologic tau in AD and reduced caudate dopamine transporter in LBD, and revealed previously unidentified findings including increased hippocampal CD47 and lowered DJ1 in AD and higher ApoE in AD with dementia. Our results were independently supported by multiplex ion beam imaging of intact tissue. This highlights the higher depth and breadth of insight on neurodegenerative diseases obtainable through SynTOF.
Collapse
Affiliation(s)
- Thanaphong Phongpreecha
- Department of Pathology, Stanford University, Stanford, CA, USA
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | | | - Candace C. Liu
- Department of Pathology, Stanford University, Stanford, CA, USA
| | | | - Alan L. Chang
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Martin Becker
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Ramin Fallahzadeh
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | | | - Nadia Postupna
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Emily Sherfield
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Dmitry Tebaykin
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Caitlin Latimer
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Carol A. Shively
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Thomas C. Register
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Suzanne Craft
- Department of Internal Medicine–Geriatrics, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | | - Edward J. Fox
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Kathleen L. Poston
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - C. Dirk Keene
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Michael Angelo
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Sean C. Bendall
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Thomas J. Montine
- Department of Pathology, Stanford University, Stanford, CA, USA
- Corresponding author.
| |
Collapse
|
7
|
Sun X, Wang X, Zhou HC, Zheng J, Su YX, Luo F. β3-adrenoceptor activation exhibits a dual effect on behaviors and glutamate receptor function in the prefrontal cortex. Behav Brain Res 2021; 412:113417. [PMID: 34157371 DOI: 10.1016/j.bbr.2021.113417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/10/2021] [Accepted: 06/14/2021] [Indexed: 11/26/2022]
Abstract
β-adrenoceptor (β-AR), especially the β1- and β2-AR subtypes, is known to participate in stress-related behavioral changes. Recently, SR58611A, a brain-penetrant β3-AR agonist, exhibits anxiolytic- and antidepressant-like effects. In this study, we sought to study the role of SR58611A in behavioral changes and its potential cellular and molecular mechanism in the prefrontal cortex (PFC). We found that rats with SR58611A (1 mg/kg) enhanced PFC-mediated recognition memory, whereas administration of higher dosage of SR58611A (20 mg/kg) caused hyperlocomotion, and exhibited an impairment effect on recognition memory. Electrophysiological data also indicated that SR58611A (1 mg/kg) selectively enhanced NMDA receptor-mediated excitatory postsynaptic currents (EPSC) through interacting with norepinephrine (NE) system and activating β3-AR, whereas higher dosage of SR58611A (20 mg/kg) reduced both AMPA receptor- and NMDA receptor-mediated EPSC. SR58611A-induced different effects on EPSC linked with the change of the surface expression quantity of NMDA receptor and/or AMPA receptor subunits. Synaptosomal-associated protein 25 (SNAP-25), which is a key soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) protein involved in incorporation of NMDA receptor to postsynaptic membrane, contributed to SR58611A (1 mg/kg)-induced enhancement of recognition memory and NMDA receptor function. Moreover, SR58611A (1 mg/kg) could rescue repeated stress-induced defect of both recognition memory and NMDA receptor function through a SNAP-25-dependent mechanism. These results provide a potential mechanism underlying the cognitive-enhancing effects of SR58611A (1 mg/kg).
Collapse
Affiliation(s)
- Xuan Sun
- School of Life Sciences, Nanchang University, Nanchang, 330031, China
| | - Xing Wang
- School of Life Sciences, Nanchang University, Nanchang, 330031, China
| | - Hou-Cheng Zhou
- Institute of Neurobiology & State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Jian Zheng
- School of Life Sciences, Nanchang University, Nanchang, 330031, China
| | - Yun-Xiao Su
- School of Life Sciences, Nanchang University, Nanchang, 330031, China
| | - Fei Luo
- School of Life Sciences, Nanchang University, Nanchang, 330031, China.
| |
Collapse
|
8
|
Zhu J, Chen F, Luo L, Wu W, Dai J, Zhong J, Lin X, Chai C, Ding P, Liang L, Wang S, Ding X, Chen Y, Wang H, Qiu J, Wang F, Sun C, Zeng Y, Fang J, Jiang X, Liu P, Tang G, Qiu X, Zhang X, Ruan Y, Jiang S, Li J, Zhu S, Xu X, Li F, Liu Z, Cao G, Chen D. Single-cell atlas of domestic pig cerebral cortex and hypothalamus. Sci Bull (Beijing) 2021; 66:1448-1461. [PMID: 36654371 DOI: 10.1016/j.scib.2021.04.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/07/2020] [Accepted: 03/12/2021] [Indexed: 01/20/2023]
Abstract
The brain of the domestic pig (Sus scrofa domesticus) has drawn considerable attention due to its high similarities to that of humans. However, the cellular compositions of the pig brain (PB) remain elusive. Here we investigated the single-nucleus transcriptomic profiles of five regions of the PB (frontal lobe, parietal lobe, temporal lobe, occipital lobe, and hypothalamus) and identified 21 cell subpopulations. The cross-species comparison of mouse and pig hypothalamus revealed the shared and specific gene expression patterns at the single-cell resolution. Furthermore, we identified cell types and molecular pathways closely associated with neurological disorders, bridging the gap between gene mutations and pathogenesis. We reported, to our knowledge, the first single-cell atlas of domestic pig cerebral cortex and hypothalamus combined with a comprehensive analysis across species, providing extensive resources for future research regarding neural science, evolutionary developmental biology, and regenerative medicine.
Collapse
Affiliation(s)
- Jiacheng Zhu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Fang Chen
- BGI-Shenzhen, Shenzhen 518083, China; MGI, BGI-Shenzhen, Shenzhen 518083, China
| | - Lihua Luo
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Weiying Wu
- BGI-Shenzhen, Shenzhen 518083, China; Department of Neurobiology, NHC and CAMS Key Laboratory of Medical Neurobiology, School of Brain Science and Brian Medicine, and the MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou 310031, China
| | - Jinxia Dai
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Jixing Zhong
- School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Xiumei Lin
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Chaochao Chai
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Peiwen Ding
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Langchao Liang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Shiyou Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Xiangning Ding
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Yin Chen
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Haoyu Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Jiaying Qiu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | | | - Chengcheng Sun
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China; School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Yuying Zeng
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China; College of Life Science, South China Agricultural University, Guangzhou 510642, China
| | - Jian Fang
- BGI-Shenzhen, Shenzhen 518083, China
| | - Xiaosen Jiang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Ping Liu
- BGI-Shenzhen, Shenzhen 518083, China; MGI, BGI-Shenzhen, Shenzhen 518083, China
| | - Gen Tang
- Shenzhen Children's Hospital, Shenzhen 518083, China
| | - Xin Qiu
- Shenzhen Children's Hospital, Shenzhen 518083, China
| | | | - Yetian Ruan
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | | | | | - Shida Zhu
- BGI-Shenzhen, Shenzhen 518083, China
| | - Xun Xu
- BGI-Shenzhen, Shenzhen 518083, China
| | - Fang Li
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Zhongmin Liu
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Gang Cao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; College of Biomedicine and Health, Huazhong Agricultural University, Wuhan 430070, China.
| | | |
Collapse
|
9
|
Mishima T, Fujiwara T, Kofuji T, Saito A, Terao Y, Akagawa K. Syntaxin 1B regulates synaptic GABA release and extracellular GABA concentration, and is associated with temperature-dependent seizures. J Neurochem 2020; 156:604-613. [PMID: 32858780 DOI: 10.1111/jnc.15159] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/13/2020] [Accepted: 08/11/2020] [Indexed: 11/29/2022]
Abstract
De novo heterozygous mutations in the STX1B gene, encoding syntaxin 1B, cause a familial, fever-associated epilepsy syndrome. Syntaxin 1B is an essential component of the pre-synaptic neurotransmitter release machinery as a soluble N-ethylmaleimide-sensitive factor attachment protein receptor protein that regulates the exocytosis of synaptic vesicles. It is also involved in regulating the functions of the SLC6 family of neurotransmitter transporters that reuptake neurotransmitters, including inhibitory neurotransmitters, such as γ-aminobutyric acid (GABA) and glycine. The purpose of the present study was to elucidate the molecular mechanisms underlying the development of febrile seizures by examining the effects of syntaxin 1B haploinsufficiency on inhibitory synaptic transmission during hyperthermia in a mouse model. Stx1b gene heterozygous knockout (Stx1b+/- ) mice showed increased susceptibility to febrile seizures and drug-induced seizures. In cultured hippocampal neurons, we examined the temperature-dependent properties of neurotransmitter release and reuptake by GABA transporter-1 (GAT-1) at GABAergic neurons using whole-cell patch-clamp recordings. The rate of spontaneous quantal GABA release was reduced in Stx1b+/- mice. The hyperthermic temperature increased the tonic GABAA current in wild-type (WT) synapses, but not in Stx1b+/- synapses. In WT neurons, recurrent bursting activities were reduced in a GABA-dependent manner at hyperthermic temperature; however, this was abolished in Stx1b+/- neurons. The blockade of GAT-1 increased the tonic GABAA current and suppressed recurrent bursting activities in Stx1b+/- neurons at the hyperthermic temperature. These data suggest that functional abnormalities associated with GABA release and reuptake in the pre-synaptic terminals of GABAergic neurons may increase the excitability of the neural circuit with hyperthermia.
Collapse
Affiliation(s)
- Tatsuya Mishima
- Department of Medical Physiology, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | - Tomonori Fujiwara
- Department of Medical Physiology, Kyorin University School of Medicine, Mitaka, Tokyo, Japan.,Faculty of Health and Medical Care, Saitama Medical University, Hidaka, Saitama, Japan
| | - Takefumi Kofuji
- Department of Medical Physiology, Kyorin University School of Medicine, Mitaka, Tokyo, Japan.,Radioisotope Laboratory, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | - Ayako Saito
- Department of Medical Physiology, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | - Yasuo Terao
- Department of Medical Physiology, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | - Kimio Akagawa
- Department of Medical Physiology, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| |
Collapse
|
10
|
Tong R, Wei C, Pan L, Zhang X. Effects of dopamine on immune signaling pathway factors, phagocytosis and exocytosis in hemocytes of Litopenaeus vannamei. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 102:103473. [PMID: 31437524 DOI: 10.1016/j.dci.2019.103473] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 08/19/2019] [Accepted: 08/19/2019] [Indexed: 06/10/2023]
Abstract
Dopamine (DA) is an important neuroendocrine factor, which can act as neurotransmitter and neurohormone. In this study, we explored the immune defense mechanism in Litopenaeus vannamei with injection of dopamine at 10-7 and 10-6 mol shrimp-1, respectively. The genes expressions of dopamine receptor (DAR), G proteins (Gs, Gi, Gq), phagocytosis and exocytosis-related proteins, as well as intracellular signaling pathway factors, and immune defense parameters were measured. Results showed that mRNA expression levels of dopamine receptor D4 (D4), Gi, nuclear transcription factors and exocytosis-related proteins decreased significantly and reached the minimum at 3 h, while the genes expressions of Gs, Gq and phagocytosis-related proteins reached the highest and lowest levels at 3 h and 6 h, respectively. The second messenger synthetases increased significantly in treatment groups within 3 h. Simultaneously, the second messengers and protein kinases shared a similar trend, which were significantly elevated and reached the peak value at 3 h. Ultimately lead to the total hemocyte count (THC), proPO activity and phagocytic activity decreased significantly, reaching minimum values at 3 h, 3 h and 6 h, respectively. While PO activity showed obvious peak changes, which maximum value reached at 3 h. These results suggested that DA receptor could couple with G protein after DA injection and might regulate immunity through cAMP-PKA, DAG-PKC or CaM pathway.
Collapse
Affiliation(s)
- Ruixue Tong
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, PR China
| | - Cun Wei
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, PR China
| | - Luqing Pan
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, PR China.
| | - Xin Zhang
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, 266003, PR China
| |
Collapse
|
11
|
The SNAP-25 Protein Family. Neuroscience 2019; 420:50-71. [DOI: 10.1016/j.neuroscience.2018.09.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 08/31/2018] [Accepted: 09/14/2018] [Indexed: 01/04/2023]
|
12
|
Pawar K, Welzel G, Haynl C, Schuster S, Scheibel T. Recombinant Spider Silk and Collagen-Based Nerve Guidance Conduits Support Neuronal Cell Differentiation and Functionality in Vitro. ACS APPLIED BIO MATERIALS 2019; 2:4872-4880. [DOI: 10.1021/acsabm.9b00628] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Kiran Pawar
- Department for Biomaterials, University of Bayreuth, Prof.-Rüdiger-Bormann-Strasse 1, 95447 Bayreuth, Germany
| | | | - Christian Haynl
- Department for Biomaterials, University of Bayreuth, Prof.-Rüdiger-Bormann-Strasse 1, 95447 Bayreuth, Germany
| | | | - Thomas Scheibel
- Department for Biomaterials, University of Bayreuth, Prof.-Rüdiger-Bormann-Strasse 1, 95447 Bayreuth, Germany
| |
Collapse
|
13
|
Jiang S, Wen N, Li Z, Dube U, Del Aguila J, Budde J, Martinez R, Hsu S, Fernandez MV, Cairns NJ, Harari O, Cruchaga C, Karch CM. Integrative system biology analyses of CRISPR-edited iPSC-derived neurons and human brains reveal deficiencies of presynaptic signaling in FTLD and PSP. Transl Psychiatry 2018; 8:265. [PMID: 30546007 PMCID: PMC6293323 DOI: 10.1038/s41398-018-0319-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 11/13/2018] [Indexed: 01/12/2023] Open
Abstract
Mutations in the microtubule-associated protein tau (MAPT) gene cause autosomal dominant frontotemporal lobar degeneration with tau inclusions (FTLD-tau). MAPT p.R406W carriers present clinically with progressive memory loss and neuropathologically with neuronal and glial tauopathy. However, the pathogenic events triggered by the expression of the mutant tau protein remain poorly understood. To identify the genes and pathways that are dysregulated in FTLD-tau, we performed transcriptomic analyses in induced pluripotent stem cell (iPSC)-derived neurons carrying MAPT p.R406W and CRISPR/Cas9-corrected isogenic controls. We found that the expression of the MAPT p.R406W mutation was sufficient to create a significantly different transcriptomic profile compared with that of the isogeneic controls and to cause the differential expression of 328 genes. Sixty-one of these genes were also differentially expressed in the same direction between MAPT p.R406W carriers and pathology-free human control brains. We found that genes differentially expressed in the stem cell models and human brains were enriched for pathways involving gamma-aminobutyric acid (GABA) receptors and pre-synaptic function. The expression of GABA receptor genes, including GABRB2 and GABRG2, were consistently reduced in iPSC-derived neurons and brains from MAPT p.R406W carriers. Interestingly, we found that GABA receptor genes, including GABRB2 and GABRG2, are significantly lower in symptomatic mouse models of tauopathy, as well as in brains with progressive supranuclear palsy. Genome wide association analyses reveal that common variants within GABRB2 are associated with increased risk for frontotemporal dementia (P < 1 × 10-3). Thus, our systems biology approach, which leverages molecular data from stem cells, animal models, and human brain tissue can reveal novel disease mechanisms. Here, we demonstrate that MAPT p.R406W is sufficient to induce changes in GABA-mediated signaling and synaptic function, which may contribute to the pathogenesis of FTLD-tau and other primary tauopathies.
Collapse
Affiliation(s)
- Shan Jiang
- Department of Psychiatry, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8134, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8111, St. Louis, MO, 63110, USA
| | - Natalie Wen
- Department of Psychiatry, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8134, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8111, St. Louis, MO, 63110, USA
| | - Zeran Li
- Department of Psychiatry, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8134, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8111, St. Louis, MO, 63110, USA
| | - Umber Dube
- Department of Psychiatry, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8134, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8111, St. Louis, MO, 63110, USA
| | - Jorge Del Aguila
- Department of Psychiatry, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8134, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8111, St. Louis, MO, 63110, USA
| | - John Budde
- Department of Psychiatry, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8134, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8111, St. Louis, MO, 63110, USA
| | - Rita Martinez
- Department of Psychiatry, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8134, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8111, St. Louis, MO, 63110, USA
| | - Simon Hsu
- Department of Psychiatry, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8134, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8111, St. Louis, MO, 63110, USA
| | - Maria V Fernandez
- Department of Psychiatry, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8134, St. Louis, MO, 63110, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8111, St. Louis, MO, 63110, USA
| | - Nigel J Cairns
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, 660S. Euclid Ave, Campus Box 8118, Saint Louis, MO, 63110, USA
| | - Oscar Harari
- Department of Psychiatry, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8134, St. Louis, MO, 63110, USA.
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8111, St. Louis, MO, 63110, USA.
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8134, St. Louis, MO, 63110, USA.
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8111, St. Louis, MO, 63110, USA.
| | - Celeste M Karch
- Department of Psychiatry, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8134, St. Louis, MO, 63110, USA.
- Hope Center for Neurological Disorders, Washington University School of Medicine, 660S. Euclid Ave. Campus Box 8111, St. Louis, MO, 63110, USA.
| |
Collapse
|
14
|
Duan MJ, Yan ML, Wang Q, Mao M, Su D, Sun LL, Li KX, Qu Y, Sun Q, Zhang XY, Huang SY, Ma JC, Ban T, Ai J. Overexpression of miR-1 in the heart attenuates hippocampal synaptic vesicle exocytosis by the posttranscriptional regulation of SNAP-25 through the transportation of exosomes. Cell Commun Signal 2018; 16:91. [PMID: 30497498 PMCID: PMC6267908 DOI: 10.1186/s12964-018-0303-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 11/13/2018] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND The link between cardiac diseases and cognitive deterioration has been accepted from the concept of "cardiogenic dementia", which was proposed in the late 1970s. However, the molecular mechanism is unclarified. METHODS The two animal models used in this study were cardiac-specific overexpression of microRNA-1-2 transgenic (Tg) mice and a myocardial infarction mouse model generated by left coronary artery ligation (LCA). First, we observed the microRNA-1 (miR-1) level and synaptic vesicles (SV) distribution in the hippocampus using in situ hybridization and transmission electron microscopy (TEM) and evaluated the expression of vesicle exocytosis related proteins by western blotting. Second, we used dual luciferase reporter assay as well as antagonist and miRNA-masking techniques to identify the posttranscriptional regulatory effect of miR-1 on the Snap25 gene. Third, FM1-43 staining was performed to investigate the effect of miR-1 on synaptic vesicle exocytosis. Lastly, we used GW4869 to inhibit the biogenesis and secretion of exosomes to determine the transportation effect of exosomes for miR-1 from the heart to the brain. RESULTS Compared with the levels in age-matched WT mice, miR-1 levels were increased in both the hearts and hippocampi of Tg mice, accompanied by the redistribution of SVs and the reduction in SV exocytosis-related protein SNAP-25 expression. In vitro studies showed that SNAP-25 protein expression was down- or upregulated by miR-1 overexpression or inhibition, respectively, however, unchanged by miRNA-masking the 3'UTR of the Snap25 gene. SV exocytosis was inhibited by miR-1 overexpression, which could be prevented by co-transfection with an anti-miR-1 oligonucleotide fragment (AMO-1). The knockdown of miR-1 by hippocampal stereotaxic injection of AMO-1 carried by a lentivirus vector (lenti-pre-AMO-1) led to the upregulation of SNAP-25 expression and prevented SV concentration in the synapses in the hippocampi of Tg mice. The application of GW4869 significantly reversed the increased miR-1 level in the blood and hippocampi as well as reduced the SNAP-25 protein levels in the hippocampi of both Tg and LCA mice. CONCLUSION The overexpression of miR-1 in the heart attenuated SV exocytosis in the hippocampus by posttranscriptionally regulating SNAP-25 through the transportation of exosomes. This study contributes to the understanding of the relationship between cardiovascular disease and brain dysfunction.
Collapse
Affiliation(s)
- Ming-Jing Duan
- Department of Pharmacology, College of Pharmacy of Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, 150086 Heilongjiang Province China
| | - Mei-Ling Yan
- Department of Pharmacology, College of Pharmacy of Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, 150086 Heilongjiang Province China
| | - Qin Wang
- Department of Pharmacology, College of Pharmacy of Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, 150086 Heilongjiang Province China
| | - Meng Mao
- Department of Pharmacology, College of Pharmacy of Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, 150086 Heilongjiang Province China
| | - Dan Su
- Department of Pharmacology, College of Pharmacy of Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, 150086 Heilongjiang Province China
| | - Lin-Lin Sun
- Department of Pharmacology, College of Pharmacy of Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, 150086 Heilongjiang Province China
| | - Ke-Xin Li
- Department of Pharmacology, College of Pharmacy of Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, 150086 Heilongjiang Province China
| | - Yang Qu
- Department of Pharmacology, College of Pharmacy of Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, 150086 Heilongjiang Province China
| | - Qiang Sun
- Department of Pharmacology, College of Pharmacy of Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, 150086 Heilongjiang Province China
| | - Xin-Yu Zhang
- Department of Pharmacology, College of Pharmacy of Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, 150086 Heilongjiang Province China
| | - Si-Yu Huang
- Department of Pharmacology, College of Pharmacy of Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, 150086 Heilongjiang Province China
| | - Ji-Chao Ma
- Department of Pharmacology, College of Pharmacy of Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, 150086 Heilongjiang Province China
| | - Tao Ban
- Department of Pharmacology, College of Pharmacy of Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, 150086 Heilongjiang Province China
| | - Jing Ai
- Department of Pharmacology, College of Pharmacy of Harbin Medical University (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin, 150086 Heilongjiang Province China
| |
Collapse
|
15
|
Pozzi D, Corradini I, Matteoli M. The Control of Neuronal Calcium Homeostasis by SNAP-25 and its Impact on Neurotransmitter Release. Neuroscience 2018; 420:72-78. [PMID: 30476527 DOI: 10.1016/j.neuroscience.2018.11.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 11/05/2018] [Accepted: 11/08/2018] [Indexed: 01/14/2023]
Abstract
The process of neurotransmitter release is central to the control of cell-to-cell communication in brain. SNAP-25 is a component of the SNARE complex, which, together with syntaxin-1 and synaptobrevin, mediates synaptic vesicle fusion with the plasma membrane. The genetic ablation of the protein or its proteolytic cleavage by botulinum neurotoxins results in a complete block of synaptic transmission. In the last years, several evidences have indicated that SNAP-25 also plays additional modulatory roles in neurotransmission through the control of voltage-gated calcium channels and presynaptic calcium ion concentration. Consistently, reduced levels of the protein affect presynaptic calcium homeostasis and result in pathologically enhanced glutamate exocytosis. The SNAP-25-dependent alterations of synaptic calcium dynamics may have direct impact on the development of neuropsychiatric disorders where the Snap-25 gene has been found to be involved.
Collapse
Affiliation(s)
- Davide Pozzi
- Humanitas University, Via Rita Levi Montalcini, 4, 20090 Pieve Emanuele, Milano, Italy; IRCCS Humanitas, via Manzoni 56, 20089 Rozzano, Italy.
| | - Irene Corradini
- CNR Institute of Neuroscience, via Vanvitelli 32, 20129 Milano, Italy
| | - Michela Matteoli
- Humanitas University, Via Rita Levi Montalcini, 4, 20090 Pieve Emanuele, Milano, Italy; IRCCS Humanitas, via Manzoni 56, 20089 Rozzano, Italy.
| |
Collapse
|
16
|
Hussain S, Ringsevjen H, Schupp M, Hvalby Ø, Sørensen JB, Jensen V, Davanger S. A possible postsynaptic role for SNAP-25 in hippocampal synapses. Brain Struct Funct 2018; 224:521-532. [PMID: 30377802 DOI: 10.1007/s00429-018-1782-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 10/19/2018] [Indexed: 12/17/2022]
Abstract
The SNARE protein SNAP-25 is well documented as regulator of presynaptic vesicle exocytosis. Increasing evidence suggests roles for SNARE proteins in postsynaptic trafficking of glutamate receptors as a basic mechanism in synaptic plasticity. Despite these indications, detailed quantitative subsynaptic localization studies of SNAP-25 have never been performed. Here, we provide novel electron microscopic data of SNAP-25 localization in postsynaptic spines. In addition to its expected presynaptic localization, we show that the protein is also present in the postsynaptic density (PSD), the postsynaptic lateral membrane and on small vesicles in the postsynaptic cytoplasm. We further investigated possible changes in synaptic SNAP-25 protein expression after hippocampal long-term potentiation (LTP). Quantitative analysis of immunogold-labeled electron microscopy sections did not show statistically significant changes of SNAP-25 gold particle densities 1 h after LTP induction, indicating that local trafficking of SNAP-25 does not play a role in the early phases of LTP. However, the strong expression of SNAP-25 in postsynaptic plasma membranes suggests a function of the protein in postsynaptic vesicle exocytosis and a possible role in hippocampal synaptic plasticity.
Collapse
Affiliation(s)
- S Hussain
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - H Ringsevjen
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - M Schupp
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ø Hvalby
- Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - J B Sørensen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - V Jensen
- Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - S Davanger
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway. .,Laboratory of Synaptic Plasticity, Division of Anatomy, Institute of Basic Medical Sciences, P.O.Box 1105, Blindern, 0317, Oslo, Norway.
| |
Collapse
|
17
|
Monteggia LM, Lin PY, Adachi M, Kavalali ET. Behavioral Analysis of SNAP-25 and Synaptobrevin-2 Haploinsufficiency in Mice. Neuroscience 2018; 420:129-135. [PMID: 30144509 DOI: 10.1016/j.neuroscience.2018.08.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 08/11/2018] [Accepted: 08/13/2018] [Indexed: 10/28/2022]
Abstract
In central synapses, synaptobrevin-2 (also called VAMP-2) is the predominant synaptic vesicle SNARE protein that interacts with the plasma membrane SNAREs, SNAP-25 and syntaxin-1 to execute exocytosis. Mice deficient in synaptobrevin-2 or SNAP-25 show embryonic lethality, which precludes investigation of the complete loss-of-function of these proteins in the adult nervous system. However, mice that carry heterozygous null mutations survive into adulthood and are fertile. In order to elucidate how loss-of-function mutations in these proteins may result in human disease phenotypes it is important to develop bona fide animal models. Therefore, given the importance of these two critical SNAREs in central synaptic transmission and their association with several neurological or neuropsychiatric disorders, we performed a comprehensive behavioral analysis of SNAP-25 heterozygous null (SNAP-25+/-) mice as well as the synaptobrevin-2 heterozygous null (+/-) mice. This analysis revealed only mild phenotypes, SNAP-25 (+/-) mice exhibited marked hypoactivity, whereas synaptobrevin-2 (+/-) mice showed enhanced performance on the rotarod. The two mouse lines did not manifest significant deficits in anxiety-related behaviors, learning and memory measures, or prepulse inhibition. The rather mild behavioral deficits indicate that these key proteins, SNAP25 and synaptobrevin-2, are expressed in excess to circumvent the impact of potential fluctuations in expression levels on nervous system function.
Collapse
Affiliation(s)
- Lisa M Monteggia
- Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA.
| | - Pei-Yi Lin
- Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Megumi Adachi
- Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | - Ege T Kavalali
- Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA.
| |
Collapse
|
18
|
Stallings NR, O'Neal MA, Hu J, Kavalali ET, Bezprozvanny I, Malter JS. Pin1 mediates Aβ 42-induced dendritic spine loss. Sci Signal 2018; 11:11/522/eaap8734. [PMID: 29559586 PMCID: PMC6136423 DOI: 10.1126/scisignal.aap8734] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Early-stage Alzheimer's disease is characterized by the loss of dendritic spines in the neocortex of the brain. This phenomenon precedes tau pathology, plaque formation, and neurodegeneration and likely contributes to synaptic loss, memory impairment, and behavioral changes in patients. Studies suggest that dendritic spine loss is induced by soluble, multimeric amyloid-β (Aβ42), which, through postsynaptic signaling, activates the protein phosphatase calcineurin. We investigated how calcineurin caused spine pathology and found that the cis-trans prolyl isomerase Pin1 was a critical downstream target of Aβ42-calcineurin signaling. In dendritic spines, Pin1 interacted with and was dephosphorylated by calcineurin, which rapidly suppressed its isomerase activity. Knockout of Pin1 or exposure to Aβ42 induced the loss of mature dendritic spines, which was prevented by exogenous Pin1. The calcineurin inhibitor FK506 blocked dendritic spine loss in Aβ42-treated wild-type cells but had no effect on Pin1-null neurons. These data implicate Pin1 in dendritic spine maintenance and synaptic loss in early Alzheimer's disease.
Collapse
Affiliation(s)
- Nancy R Stallings
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Melissa A O'Neal
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Jie Hu
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Ege T Kavalali
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ilya Bezprozvanny
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - James S Malter
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA.
| |
Collapse
|
19
|
Williams CL, Smith SM. Calcium dependence of spontaneous neurotransmitter release. J Neurosci Res 2018; 96:335-347. [PMID: 28699241 PMCID: PMC5766384 DOI: 10.1002/jnr.24116] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 06/16/2017] [Accepted: 06/19/2017] [Indexed: 01/14/2023]
Abstract
Spontaneous release of neurotransmitters is regulated by extracellular [Ca2+ ] and intracellular [Ca2+ ]. Curiously, some of the mechanisms of Ca2+ signaling at central synapses are different at excitatory and inhibitory synapses. While the stochastic activity of voltage-activated Ca2+ channels triggers a majority of spontaneous release at inhibitory synapses, this is not the case at excitatory nerve terminals. Ca2+ release from intracellular stores regulates spontaneous release at excitatory and inhibitory terminals, as do agonists of the Ca2+ -sensing receptor. Molecular machinery triggering spontaneous vesicle fusion may differ from that underlying evoked release and may be one of the sources of heterogeneity in release mechanisms.
Collapse
Affiliation(s)
- Courtney L. Williams
- Department of Medicine, Division of Pulmonary & Critical Care Medicine, Oregon Health & Science University, Portland, Oregon, 97239, USA
- Section of Pulmonary & Critical Care Medicine, VA Portland Health Care System, Portland, Oregon, USA
| | - Stephen M. Smith
- Department of Medicine, Division of Pulmonary & Critical Care Medicine, Oregon Health & Science University, Portland, Oregon, 97239, USA
- Section of Pulmonary & Critical Care Medicine, VA Portland Health Care System, Portland, Oregon, USA
| |
Collapse
|
20
|
Dunn AR, Hoffman CA, Stout KA, Ozawa M, Dhamsania RK, Miller GW. Immunochemical analysis of the expression of SV2C in mouse, macaque and human brain. Brain Res 2017; 1702:85-95. [PMID: 29274878 DOI: 10.1016/j.brainres.2017.12.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 11/30/2017] [Accepted: 12/19/2017] [Indexed: 11/17/2022]
Abstract
The synaptic vesicle glycoprotein 2C (SV2C) is an undercharacterized protein with enriched expression in phylogenetically old brain regions. Its precise role within the brain is unclear, though various lines of evidence suggest that SV2C is involved in the function of synaptic vesicles through the regulation of vesicular trafficking, calcium-induced exocytosis, or synaptotagmin function. SV2C has been linked to multiple neurological disorders, including Parkinson's disease and psychiatric conditions. SV2C is expressed in various cell types-primarily dopaminergic, GABAergic, and cholinergic cells. In mice, it is most highly expressed in nuclei within the basal ganglia, though it is unknown if this pattern of expression is consistent across species. Here, we use a custom SV2C-specific antiserum to describe localization within the brain of mouse, nonhuman primate, and human, including cell-type localization. We found that the immunoreactivity with this antiserum is consistent with previously-published antibodies, and confirmed localization of SV2C in the basal ganglia of rodent, rhesus macaque, and human. We observed strongest expression of SV2C in the substantia nigra, ventral tegmental area, dorsal striatum, pallidum, and nucleus accumbens of each species. Further, we demonstrate colocalization between SV2C and markers of dopaminergic, GABAergic, and cholinergic neurons within these brain regions. SV2C has been increasingly linked to dopamine and basal ganglia function. These antisera will be an important resource moving forward in our understanding of the role of SV2C in vesicle dynamics and neurological disease.
Collapse
Affiliation(s)
- Amy R Dunn
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Carlie A Hoffman
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Kristen A Stout
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Minagi Ozawa
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Rohan K Dhamsania
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Gary W Miller
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States; Center for Neurodegenerative Disease, Emory University, Atlanta, GA 30322, United States.
| |
Collapse
|
21
|
Kenny K, Royer L, Moore AR, Chen X, Marr MT, Paradis S. Rem2 signaling affects neuronal structure and function in part by regulation of gene expression. Mol Cell Neurosci 2017; 85:190-201. [PMID: 29066292 DOI: 10.1016/j.mcn.2017.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 09/27/2017] [Accepted: 10/11/2017] [Indexed: 12/12/2022] Open
Abstract
The central nervous system has the remarkable ability to convert changes in the environment in the form of sensory experience into long-term alterations in synaptic connections and dendritic arborization, in part through changes in gene expression. Surprisingly, the molecular mechanisms that translate neuronal activity into changes in neuronal connectivity and morphology remain elusive. Rem2, a member of the Rad/Rem/Rem2/Gem/Kir (RGK) subfamily of small Ras-like GTPases, is a positive regulator of synapse formation and negative regulator of dendritic arborization. Here we identify that one output of Rem2 signaling is the regulation of gene expression. Specifically, we demonstrate that Rem2 signaling modulates the expression of genes required for a variety of cellular processes from neurite extension to synapse formation and synaptic function. Our results highlight Rem2 as a unique molecule that transduces changes in neuronal activity detected at the cell membrane to morphologically relevant changes in gene expression in the nucleus.
Collapse
Affiliation(s)
- Katelyn Kenny
- Department of Biology, Brandeis University, Waltham, MA 02454, United States
| | - Leandro Royer
- Department of Biology, Brandeis University, Waltham, MA 02454, United States
| | - Anna R Moore
- Department of Biology, Brandeis University, Waltham, MA 02454, United States; Volen Center for Complex Systems, Brandeis University, Waltham, MA 02454, United States; National Center for Behavioral Genomics, Brandeis University, Waltham, MA 02454, United States
| | - Xiao Chen
- Department of Biology, Brandeis University, Waltham, MA 02454, United States; National Center for Behavioral Genomics, Brandeis University, Waltham, MA 02454, United States
| | - Michael T Marr
- Department of Biology, Brandeis University, Waltham, MA 02454, United States; Rosenstiel Basic Medical Sciences Research Center, Brandeis University, Waltham, MA 02454, United States
| | - Suzanne Paradis
- Department of Biology, Brandeis University, Waltham, MA 02454, United States; Volen Center for Complex Systems, Brandeis University, Waltham, MA 02454, United States; National Center for Behavioral Genomics, Brandeis University, Waltham, MA 02454, United States.
| |
Collapse
|
22
|
Vaes N, Lentjes MHFM, Gijbels MJ, Rademakers G, Daenen KL, Boesmans W, Wouters KAD, Geuzens A, Qu X, Steinbusch HPJ, Rutten BPF, Baldwin SH, Sharkey KA, Hofstra RMW, van Engeland M, Vanden Berghe P, Melotte V. NDRG4, an early detection marker for colorectal cancer, is specifically expressed in enteric neurons. Neurogastroenterol Motil 2017; 29. [PMID: 28524415 DOI: 10.1111/nmo.13095] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 03/30/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND Promoter methylation of N-myc Downstream-Regulated Gene 4 (NDRG4) in fecal DNA is an established early detection marker for colorectal cancer (CRC). Despite its connection to CRC, NDRG4 is predominantly studied in brain and heart, with little to no knowledge about its expression or role in other organs. In this study, we aimed to determine the whole-body expression of NDRG4, with a focus on the intestinal tract. METHODS We investigated NDRG4 expression throughout the body by immunohistochemistry, Western Blotting and in situ mRNA hybridization using tissues from NDRG4 wild-type, heterozygous and knockout mice and humans. In addition, we explored cell-specific expression of NDRG4 in murine whole-mount gut preparations using immunofluorescence and confocal microscopy. KEY RESULTS NDRG4 is specifically expressed within nervous system structures throughout the body. In the intestinal tract of both mouse and man, NDRG4 immunoreactivity was restricted to the enteric nervous system (ENS), where it labeled cell bodies of the myenteric and submucosal plexuses and interconnecting nerve fibers. More precisely, NDRG4 expression was limited to neurons, as NDRG4 always co-localized with HuC/D (pan-neuronal marker) but never with GFAP (an enteric glial cell marker). Furthermore, NDRG4 was expressed in various neuropeptide Y positive neurons, but was only found in a minority (~10%) of neurons expressing neuronal nitric oxide synthase. CONCLUSIONS AND INFERENCES NDRG4 is exclusively expressed by central, peripheral and enteric neurons/nerves, suggesting a neuronal-specific role of this protein. Our findings raise the question whether NDRG4, via the ENS, an understudied component of the tumor microenvironment, supports CRC development and/or progression.
Collapse
Affiliation(s)
- N Vaes
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - M H F M Lentjes
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - M J Gijbels
- Departments of Pathology and Molecular Genetics, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - G Rademakers
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - K L Daenen
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - W Boesmans
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands.,Laboratory for Enteric Neuroscience (LENS); Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium
| | - K A D Wouters
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - A Geuzens
- Laboratory for Enteric Neuroscience (LENS); Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium
| | - X Qu
- Department of Pediatric Cardiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - H P J Steinbusch
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University Medical Centre, Maastricht, The Netherlands
| | - B P F Rutten
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University Medical Centre, Maastricht, The Netherlands
| | - S H Baldwin
- Department of Pediatric Cardiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - K A Sharkey
- Hotchkiss Brain Institute and Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - R M W Hofstra
- Department of Clinical Genetics, University of Rotterdam, EMC, Rotterdam, The Netherlands
| | - M van Engeland
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - P Vanden Berghe
- Laboratory for Enteric Neuroscience (LENS); Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium
| | - V Melotte
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands.,Department of Clinical Genetics, University of Rotterdam, EMC, Rotterdam, The Netherlands
| |
Collapse
|
23
|
Münster-Wandowski A, Heilmann H, Bolduan F, Trimbuch T, Yanagawa Y, Vida I. Distinct Localization of SNAP47 Protein in GABAergic and Glutamatergic Neurons in the Mouse and the Rat Hippocampus. Front Neuroanat 2017; 11:56. [PMID: 28751858 PMCID: PMC5508021 DOI: 10.3389/fnana.2017.00056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 06/22/2017] [Indexed: 12/14/2022] Open
Abstract
Synaptosomal-associated protein of 47 kDa (SNAP47) isoform is an atypical member of the SNAP family, which does not contribute directly to exocytosis and synaptic vesicle (SV) recycling. Initial characterization of SNAP47 revealed a widespread expression in nervous tissue, but little is known about its cellular and subcellular localization in hippocampal neurons. Therefore, in the present study we applied multiple-immunofluorescence labeling, immuno-electron microscopy and in situ hybridization (ISH) and analyzed the localization of SNAP47 in pre- and postsynaptic compartments of glutamatergic and GABAergic neurons in the mouse and rat hippocampus. While the immunofluorescence signal for SNAP47 showed a widespread distribution in both mouse and rat, the labeling pattern was complementary in the two species: in the mouse the immunolabeling was higher over the CA3 stratum radiatum, oriens and cell body layer. In contrast, in the rat the labeling was stronger over the CA1 neuropil and in the CA3 stratum lucidum. Furthermore, in the mouse high somatic labeling for SNAP47 was observed in GABAergic interneurons (INs). On the contrary, in the rat, while most INs were positive, they blended in with the high neuropil labeling. ISH confirmed the high expression of SNAP47 RNA in INs in the mouse. Co-staining for SNAP47 and pre- and postsynaptic markers in the rat revealed a strong co-localization postsynaptically with PSD95 in dendritic spines of pyramidal cells and, to a lesser extent, presynaptically, with ZnT3 and vesicular glutamate transporter 1 (VGLUT1) in glutamatergic terminals such as mossy fiber (MF) boutons. Ultrastructural analysis confirmed the pre- and postsynaptic localization at glutamatergic synapses. Furthermore, in the mouse hippocampus SNAP47 was found to be localized at low levels to dendritic shafts and axon terminals of putative INs forming symmetric synapses, indicating that this protein could be trafficked to both post- and presynaptic sites in both major cell types. These results reveal divergent localization of SNAP47 protein in mouse and rat hippocampus indicating species- and cell type-specific differences. SNAP47 is likely to be involved in unique fusion machinery which is distinct from the one involved in presynaptic neurotransmitter release. Nonetheless, our data suggest that SNAP47 may be involved not only postsynaptic, but also in presynaptic function.
Collapse
Affiliation(s)
| | - Heike Heilmann
- Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin BerlinBerlin, Germany
| | - Felix Bolduan
- Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin BerlinBerlin, Germany
| | - Thorsten Trimbuch
- Institute of Neurophysiology, Charité-Universitätsmedizin BerlinBerlin, Germany
| | - Yuchio Yanagawa
- Departments of Genetic and Behavioral Neuroscience, Gunma University, Graduate School of MedicineMaebashi City, Japan
| | - Imre Vida
- Institute of Integrative Neuroanatomy, Charité-Universitätsmedizin BerlinBerlin, Germany.,Neurocure Cluster of Excellence, Charité-Universitätsmedizin BerlinBerlin, Germany
| |
Collapse
|
24
|
Matsuno T, Kiyokage E, Toida K. Synaptic distribution of individually labeled mitral cells in the external plexiform layer of the mouse olfactory bulb. J Comp Neurol 2017; 525:1633-1648. [PMID: 27864926 DOI: 10.1002/cne.24148] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 06/28/2016] [Accepted: 10/11/2016] [Indexed: 11/07/2022]
Abstract
Mitral cells are the major projection neurons of the olfactory bulb. They receive olfactory inputs, regulate information, and project their axons to the olfactory cortex. To understand output regulation of mitral cells better, we established a method to visualize individual projection neurons and quantitatively examined their synaptic distribution. Individual mitral cells were labeled by viral injection, reconstructed three dimensionally with light microscopy, and serial sectioned for electron microscopy. Synaptic distributions were analyzed in electron microscopically reconstructed cell bodies, two regions of secondary dendrites (near the somata and ∼200 μm from the somata), and primary dendrites. The ratio of presynaptic sites (60%) and reciprocal synapses (60% presynaptic and 80% postsynaptic sites) were similar in each region. Characteristically, primary dendrite synapses were distributed mainly within the inner half of the external plexiform layer (EPL). For comparison, tufted cells were also examined, and the synaptic distribution in two secondary dendrite regions, which corresponded with mitral cells, was analyzed. The results showed that the ratio of reciprocal synapses (80% presynaptic and 90% postsynaptic sites) was greater than in mitral cells. The distribution of symmetrical synapses was also analyzed with synaptic and neuronal markers, such as parvalbumin, vesicular gamma-aminobutyric acid transporter, and gephyrin. Parvalbumin-expressing neurons tended to form synapses on secondary dendrites near the somata and were more uniformly distributed on primary dendrites of mitral cells. These results indicate that local mitral cell synaptic circuits are formed in accordance with their functional roles and restricted to the inner half of the EPL. J. Comp. Neurol. 525:1633-1648, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Takeshi Matsuno
- Department of Anatomy, Kawasaki Medical School, Okayama, Japan
| | - Emi Kiyokage
- Department of Anatomy, Kawasaki Medical School, Okayama, Japan
| | - Kazunori Toida
- Department of Anatomy, Kawasaki Medical School, Okayama, Japan.,Research Center for Ultra-High Voltage Electron Microscopy, Osaka University, Osaka, Japan
| |
Collapse
|
25
|
Toft-Bertelsen TL, Ziomkiewicz I, Houy S, Pinheiro PS, Sørensen JB. Regulation of Ca2+ channels by SNAP-25 via recruitment of syntaxin-1 from plasma membrane clusters. Mol Biol Cell 2016; 27:3329-3341. [PMID: 27605709 PMCID: PMC5170865 DOI: 10.1091/mbc.e16-03-0184] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 09/01/2016] [Indexed: 12/20/2022] Open
Abstract
SNAP-25 regulates Ca2+ channels in an unknown manner. Endogenous and exogenous SNAP-25 inhibit Ca2+ currents indirectly by recruiting syntaxin-1 from clusters on the plasma membrane, thereby making it available for Ca2+ current inhibition. Thus the cell can regulate Ca2+ influx by expanding or contracting syntaxin-1 clusters. SNAP-25 regulates Ca2+ channels, with potentially important consequences for diseases involving an aberrant SNAP-25 expression level. How this regulation is executed mechanistically remains unknown. We investigated this question in mouse adrenal chromaffin cells and found that SNAP-25 inhibits Ca2+ currents, with the B-isoform being more potent than the A-isoform, but not when syntaxin-1 is cleaved by botulinum neurotoxin C. In contrast, syntaxin-1 inhibits Ca2+ currents independently of SNAP-25. Further experiments using immunostaining showed that endogenous or exogenous SNAP-25 expression recruits syntaxin-1 from clusters on the plasma membrane, thereby increasing the immunoavailability of syntaxin-1 and leading indirectly to Ca2+ current inhibition. Expression of Munc18-1, which recruits syntaxin-1 within the exocytotic pathway, does not modulate Ca2+ channels, whereas overexpression of the syntaxin-binding protein Doc2B or ubMunc13-2 increases syntaxin-1 immunoavailability and concomitantly down-regulates Ca2+ currents. Similar findings were obtained upon chemical cholesterol depletion, leading directly to syntaxin-1 cluster dispersal and Ca2+ current inhibition. We conclude that clustering of syntaxin-1 allows the cell to maintain a high syntaxin-1 expression level without compromising Ca2+ influx, and recruitment of syntaxin-1 from clusters by SNAP-25 expression makes it available for regulating Ca2+ channels. This mechanism potentially allows the cell to regulate Ca2+ influx by expanding or contracting syntaxin-1 clusters.
Collapse
Affiliation(s)
- Trine Lisberg Toft-Bertelsen
- Neurosecretion Group, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Iwona Ziomkiewicz
- Neurosecretion Group, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Sébastien Houy
- Neurosecretion Group, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Paulo S Pinheiro
- Neurosecretion Group, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Jakob B Sørensen
- Neurosecretion Group, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| |
Collapse
|
26
|
Abstract
In mature neurons AMPA receptors cluster at excitatory synapses primarily on dendritic spines, whereas GABAA receptors cluster at inhibitory synapses mainly on the soma and dendritic shafts. The molecular mechanisms underlying the precise sorting of these receptors remain unclear. By directly studying the constitutive exocytic vesicles of AMPA and GABAA receptors in vitro and in vivo, we demonstrate that they are initially sorted into different vesicles in the Golgi apparatus and inserted into distinct domains of the plasma membrane. These insertions are dependent on distinct Rab GTPases and SNARE complexes. The insertion of AMPA receptors requires SNAP25-syntaxin1A/B-VAMP2 complexes, whereas insertion of GABAA receptors relies on SNAP23-syntaxin1A/B-VAMP2 complexes. These SNARE complexes affect surface targeting of AMPA or GABAA receptors and synaptic transmission. Our studies reveal vesicular sorting mechanisms controlling the constitutive exocytosis of AMPA and GABAA receptors, which are critical for the regulation of excitatory and inhibitory responses in neurons.
Collapse
|
27
|
Crawford DC, Kavalali ET. Molecular underpinnings of synaptic vesicle pool heterogeneity. Traffic 2015; 16:338-64. [PMID: 25620674 DOI: 10.1111/tra.12262] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 01/06/2015] [Indexed: 12/31/2022]
Abstract
Neuronal communication relies on chemical synaptic transmission for information transfer and processing. Chemical neurotransmission is initiated by synaptic vesicle fusion with the presynaptic active zone resulting in release of neurotransmitters. Classical models have assumed that all synaptic vesicles within a synapse have the same potential to fuse under different functional contexts. In this model, functional differences among synaptic vesicle populations are ascribed to their spatial distribution in the synapse with respect to the active zone. Emerging evidence suggests, however, that synaptic vesicles are not a homogenous population of organelles, and they possess intrinsic molecular differences and differential interaction partners. Recent studies have reported a diverse array of synaptic molecules that selectively regulate synaptic vesicles' ability to fuse synchronously and asynchronously in response to action potentials or spontaneously irrespective of action potentials. Here we discuss these molecular mediators of vesicle pool heterogeneity that are found on the synaptic vesicle membrane, on the presynaptic plasma membrane, or within the cytosol and consider some of the functional consequences of this diversity. This emerging molecular framework presents novel avenues to probe synaptic function and uncover how synaptic vesicle pools impact neuronal signaling.
Collapse
Affiliation(s)
- Devon C Crawford
- Department of Neuroscience, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-9111, USA
| | | |
Collapse
|
28
|
Thompson PM, Cruz DA, Fucich EA, Olukotun DY, Takahashi M, Itakura M. SNAP-25a/b Isoform Levels in Human Brain Dorsolateral Prefrontal Cortex and Anterior Cingulate Cortex. MOLECULAR NEUROPSYCHIATRY 2015; 1:220-34. [PMID: 27606314 DOI: 10.1159/000441224] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 09/09/2015] [Indexed: 01/03/2023]
Abstract
SNAP-25 is a neurotransmitter vesicular docking protein which has been associated with brain disorders such as attention deficit hyperactivity disorder, bipolar disorder and schizophrenia. In this project, we were interested if clinical factors are associated with differential SNAP-25 expression. We examined the SNAP-25 isoform mRNA and protein levels in postmortem cortex Brodmann's area 9 (BA9) and BA24 (n = 29). Subjects were divided by psychiatric diagnosis, clinical variables including mood state in the last week of life and lifetime impulsiveness. We found affected subjects with a diagnosis of alcohol use disorder (AUD) had a lower level of SNAP-25b BA24 protein compared to those without AUD. Hispanic subjects had lower levels of SNAP-25a, b and BA9 mRNA than Anglo-American subjects. Subjects who smoked had a total pan (total) SNAP-25 BA9/BA24 ratio. Subjects in the group with a low level of anxious-psychotic symptoms had higher SNAP-25a BA24 mRNA compared to normal controls, and both the high and low symptoms groups had higher pan (total) SNAP-25 BA9/BA24 ratios than normal controls. These data expand our understanding of clinical factors associated with SNAP-25. They suggest that SNAP-25 total and isoform levels may be useful biomarkers beyond limited neurological and psychiatric diagnostic categories.
Collapse
Affiliation(s)
| | - Dianne A Cruz
- Departments of Psychiatry, University of Texas Health Science Center San Antonio, San Antonio, Tex., USA
| | - Elizabeth A Fucich
- Departments of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, Tex., USA
| | - Dianna Y Olukotun
- Departments of Psychiatry, University of Texas Health Science Center San Antonio, San Antonio, Tex., USA
| | - Masami Takahashi
- Department of Biochemistry, Kitasato University School of Medicine, Tokyo, Japan
| | - Makoto Itakura
- Department of Biochemistry, Kitasato University School of Medicine, Tokyo, Japan
| |
Collapse
|
29
|
Lundwall RA, Dannemiller JL, Goldsmith HH. Genetic associations with reflexive visual attention in infancy and childhood. Dev Sci 2015; 20. [PMID: 26613685 DOI: 10.1111/desc.12371] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 08/28/2015] [Indexed: 12/11/2022]
Abstract
This study elucidates genetic influences on reflexive (as opposed to sustained) attention in children (aged 9-16 years; N = 332) who previously participated as infants in visual attention studies using orienting to a moving bar (Dannemiller, 2004). We investigated genetic associations with reflexive attention measures in infancy and childhood in the same group of children. The genetic markers (single nucleotide polymorphisms and variable number tandem repeats on the genes APOE, BDNF, CHRNA4, COMT, DRD4, HTR4, IGF2, MAOA, SLC5A7, SLC6A3, and SNAP25) are related to brain development and/or to the availability of neurotransmitters such as acetylcholine, dopamine, or serotonin. This study shows that typically developing children have differences in reflexive attention associated with their genes, as we found in adults (Lundwall, Guo & Dannemiller, 2012). This effort to extend our previous findings to outcomes in infancy and childhood was necessary because genetic influence may differ over the course of development. Although two of the genes that were tested in our adult study (Lundwall et al., 2012) were significant in either our infant study (SLC6A3) or child study (DRD4), the specific markers tested differed. Performance on the infant task was associated with SLC6A3. In addition, several genetic associations with an analogous child task occurred with markers on CHRNA4, COMT, and DRD4. Interestingly, the child version of the task involved an interaction such that which genotype group performed poorer on the child task depended on whether we were examining the higher or lower infant scoring group. These findings are discussed in terms of genetic influences on reflexive attention in infancy and childhood.
Collapse
|
30
|
Bragina L, Bonifacino T, Bassi S, Milanese M, Bonanno G, Conti F. Differential expression of metabotropic glutamate and GABA receptors at neocortical glutamatergic and GABAergic axon terminals. Front Cell Neurosci 2015; 9:345. [PMID: 26388733 PMCID: PMC4559644 DOI: 10.3389/fncel.2015.00345] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 08/18/2015] [Indexed: 12/04/2022] Open
Abstract
Metabotropic glutamate (Glu) receptors (mGluRs) and GABAB receptors are highly expressed at presynaptic sites. To verify the possibility that the two classes of metabotropic receptors contribute to axon terminals heterogeneity, we studied the localization of mGluR1α, mGluR5, mGluR2/3, mGluR7, and GABAB1 in VGLUT1-, VGLUT2-, and VGAT- positive terminals in the cerebral cortex of adult rats. VGLUT1-positive puncta expressed mGluR1α (∼5%), mGluR5 (∼6%), mGluR2/3 (∼22%), mGluR7 (∼17%), and GABAB1 (∼40%); VGLUT2-positive terminals expressed mGluR1α (∼10%), mGluR5 (∼11%), mGluR2/3 (∼20%), mGluR7 (∼28%), and GABAB1 (∼25%); whereas VGAT-positive puncta expressed mGluR1α (∼27%), mGluR5 (∼24%), mGluR2/3 (∼38%), mGluR7 (∼31%), and GABAB1 (∼19%). Control experiments ruled out the possibility that postsynaptic mGluRs and GABAB1 might have significantly biased our results. We also performed functional assays in synaptosomal preparations, and showed that all agonists modify Glu and GABA levels, which return to baseline upon exposure to antagonists. Overall, these findings indicate that mGluR1α, mGluR5, mGluR2/3, mGluR7, and GABAB1 expression differ significantly between glutamatergic and GABAergic axon terminals, and that the robust expression of heteroreceptors may contribute to the homeostatic regulation of the balance between excitation and inhibition.
Collapse
Affiliation(s)
- Luca Bragina
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle MarcheAncona, Italy
- Center for Neurobiology of Aging, Istituto Nazionale di Riposo e Cura per Anziani – Istituto di Ricovero e Cura a Carattere ScientificoAncona, Italy
| | - Tiziana Bonifacino
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of GenoaGenoa, Italy
| | - Silvia Bassi
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle MarcheAncona, Italy
| | - Marco Milanese
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of GenoaGenoa, Italy
| | - Giambattista Bonanno
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of GenoaGenoa, Italy
- Center of Excellence for Biomedical Research, University of GenoaGenoa, Italy
| | - Fiorenzo Conti
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle MarcheAncona, Italy
- Center for Neurobiology of Aging, Istituto Nazionale di Riposo e Cura per Anziani – Istituto di Ricovero e Cura a Carattere ScientificoAncona, Italy
- Fondazione di Medicina Molecolare, Università Politecnica delle MarcheAncona, Italy
| |
Collapse
|
31
|
Xu H, Shan XF, Cong X, Yang NY, Wu LL, Yu GY, Zhang Y, Cai ZG. Pre- and Post-synaptic Effects of Botulinum Toxin A on Submandibular Glands. J Dent Res 2015; 94:1454-62. [PMID: 26078423 DOI: 10.1177/0022034515590087] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Intraglandular injection of botulinum toxin type A (BoNT/A) is an effective treatment for sialorrhea. Despite numerous experimental and clinical studies on inhibition of saliva section by BoNT/A, the proteolysis of synaptosomal-associated protein 25 (SNAP-25) following BoNT/A treatment has not yet been confirmed in the salivary gland after injection of BoNT/A. More important, it is not known whether BoNT/A exerts a direct effect in acinar cells. Here, we show that injection of BoNT/A into the rat submandibular gland (SMG) decreased salivary flow in a dose-dependent manner; the inhibitory effect lasted at least 4 wk, and salivary flow recovered to normal levels by 12 wk. During the inhibitory period, SMG neurons and synapses expressed lower levels of full-length SNAP-25, and cleavage of SNAP-25 was observed, as indicated by detection of reduced molecular weight SNAP-25 using Western blotting. In addition, the water channel aquaporin 5 (AQP5) was downregulated and abnormally distributed in rat SMG after injection of BoNT/A. The direct effects of BoNT/A on AQP5 expression and distribution were assessed in vitro to exclude the influence of BoNT/A-induced inhibitory neurotransmission. In stable GFP-AQP5-transfected SMG-C6 cells, treatment with BoNT/A reduced the cell surface protein level of AQP5 in a dose- and time-dependent manner without affecting total AQP5 protein expression. Cell surface biotinylation and immunofluorescence demonstrated translocation of AQP5 from the membrane to the cytoplasm, which was confirmed by decreased levels of AQP5 protein in the membrane fraction and increased levels in the cytoplasmic fraction, suggestive of AQP5 redistribution. Taken together, these results indicated that BoNT/A reversibly decreased saliva secretion in rat SMGs through not only the presynaptic SNAP-25 cleavage but also the postsynaptic AQP5 redistribution. These data provide the first evidence for a direct effect of BoNT/A on the salivary gland.
Collapse
Affiliation(s)
- H Xu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| | - X F Shan
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| | - X Cong
- Department of Physiology and Pathophysiology, Peking University Health Science Centre and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - N Y Yang
- Department of Pediatric Dentistry, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - L L Wu
- Department of Physiology and Pathophysiology, Peking University Health Science Centre and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - G Y Yu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| | - Y Zhang
- Department of Physiology and Pathophysiology, Peking University Health Science Centre and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Z G Cai
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
32
|
Beske PH, Scheeler SM, Adler M, McNutt PM. Accelerated intoxication of GABAergic synapses by botulinum neurotoxin A disinhibits stem cell-derived neuron networks prior to network silencing. Front Cell Neurosci 2015; 9:159. [PMID: 25954159 PMCID: PMC4407583 DOI: 10.3389/fncel.2015.00159] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 04/08/2015] [Indexed: 11/13/2022] Open
Abstract
Botulinum neurotoxins (BoNTs) are extremely potent toxins that specifically cleave SNARE proteins in peripheral synapses, preventing neurotransmitter release. Neuronal responses to BoNT intoxication are traditionally studied by quantifying SNARE protein cleavage in vitro or monitoring physiological paralysis in vivo. Consequently, the dynamic effects of intoxication on synaptic behaviors are not well-understood. We have reported that mouse embryonic stem cell-derived neurons (ESNs) are highly sensitive to BoNT based on molecular readouts of intoxication. Here we study the time-dependent changes in synapse- and network-level behaviors following addition of BoNT/A to spontaneously active networks of glutamatergic and GABAergic ESNs. Whole-cell patch-clamp recordings indicated that BoNT/A rapidly blocked synaptic neurotransmission, confirming that ESNs replicate the functional pathophysiology responsible for clinical botulism. Quantitation of spontaneous neurotransmission in pharmacologically isolated synapses revealed accelerated silencing of GABAergic synapses compared to glutamatergic synapses, which was consistent with the selective accumulation of cleaved SNAP-25 at GAD1+ pre-synaptic terminals at early timepoints. Different latencies of intoxication resulted in complex network responses to BoNT/A addition, involving rapid disinhibition of stochastic firing followed by network silencing. Synaptic activity was found to be highly sensitive to SNAP-25 cleavage, reflecting the functional consequences of the localized cleavage of the small subpopulation of SNAP-25 that is engaged in neurotransmitter release in the nerve terminal. Collectively these findings illustrate that use of synaptic function assays in networked neurons cultures offers a novel and highly sensitive approach for mechanistic studies of toxin:neuron interactions and synaptic responses to BoNT.
Collapse
Affiliation(s)
- Phillip H Beske
- Cellular and Molecular Biology Branch, Research Division, United States Army Medical Research Institute of Chemical Defense Aberdeen Proving Ground, MD, USA
| | - Stephen M Scheeler
- Cellular and Molecular Biology Branch, Research Division, United States Army Medical Research Institute of Chemical Defense Aberdeen Proving Ground, MD, USA
| | - Michael Adler
- Cellular and Molecular Biology Branch, Research Division, United States Army Medical Research Institute of Chemical Defense Aberdeen Proving Ground, MD, USA
| | - Patrick M McNutt
- Cellular and Molecular Biology Branch, Research Division, United States Army Medical Research Institute of Chemical Defense Aberdeen Proving Ground, MD, USA
| |
Collapse
|
33
|
Abstract
Experimental advances in the study of neuroglia signaling have been greatly accelerated by the generation of transgenic mouse models. In particular, an elegant manipulation that interferes with astrocyte vesicular release of gliotransmitters via overexpression of a dominant-negative domain of vesicular SNARE (dnSNARE) has led to documented astrocytic involvement in processes that were traditionally considered strictly neuronal, including the sleep-wake cycle, LTP, cognition, cortical slow waves, depression, and pain. A key premise leading to these conclusions was that expression of the dnSNARE was specific to astrocytes. Inconsistent with this premise, we report here widespread expression of the dnSNARE transgene in cortical neurons. We further demonstrate that the activity of cortical neurons is reversibly suppressed in dnSNARE mice. These findings highlight the need for independent validation of astrocytic functions identified in dnSNARE mice and thus question critical evidence that astrocytes contribute to neurotransmission through SNARE-dependent vesicular release of gliotransmitters.
Collapse
|
34
|
Nava N, Treccani G, Liebenberg N, Chen F, Popoli M, Wegener G, Nyengaard JR. Chronic desipramine prevents acute stress-induced reorganization of medial prefrontal cortex architecture by blocking glutamate vesicle accumulation and excitatory synapse increase. Int J Neuropsychopharmacol 2015; 18:pyu085. [PMID: 25522419 PMCID: PMC4360240 DOI: 10.1093/ijnp/pyu085] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Although a clear negative influence of chronic exposure to stressful experiences has been repeatedly demonstrated, the outcome of acute stress on key brain regions has only just started to be elucidated. Although it has been proposed that acute stress may produce enhancement of brain plasticity and that antidepressants may prevent such changes, we still lack ultrastructural evidence that acute stress-induced changes in neurotransmitter physiology are coupled with structural synaptic modifications. METHODS Rats were pretreated chronically (14 days) with desipramine (10mg/kg) and then subjected to acute foot-shock stress. By means of serial section electron microscopy, the structural remodeling of medial prefrontal cortex glutamate synapses was assessed soon after acute stressor cessation and stress hormone levels were measured. RESULTS Foot-shock stress induced a remarkable increase in the number of docked vesicles and small excitatory synapses, partially and strongly prevented by desipramine pretreatment, respectively. Acute stress-induced corticosterone elevation was not affected by drug treatment. CONCLUSIONS Since desipramine pretreatment prevented the stress-induced structural plasticity but not the hormone level increase, we hypothesize that the preventing action of desipramine is located on pathways downstream of this process and/or other pathways. Moreover, because enhancement of glutamate system remodeling may contribute to overexcitation dysfunctions, this aspect could represent a crucial component in the pathophysiology of stress-related disorders.
Collapse
Affiliation(s)
- Nicoletta Nava
- Stereology and Electron Microscopy Laboratory, Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University Hospital, Aarhus, Denmark (Drs Nava, Chen, and Nyengaard); Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov, Denmark (Drs Nava, Treccani, Liebenberg, Chen, and Wegener); Pharmaceutical Research Center of Excellence, School of Pharmacy, North-West University, Potchefstroom, South Africa (Dr Wegener); Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmacologiche e Biomolecolari and Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milano, Italy (Drs Treccani and Popoli).
| | | | | | | | | | | | | |
Collapse
|
35
|
Gallart-Palau X, Serra A, Qian J, Chen CP, Kalaria RN, Sze SK. Temporal lobe proteins implicated in synaptic failure exhibit differential expression and deamidation in vascular dementia. Neurochem Int 2014; 80:87-98. [PMID: 25497727 DOI: 10.1016/j.neuint.2014.12.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 11/26/2014] [Accepted: 12/02/2014] [Indexed: 12/20/2022]
Abstract
Progressive synaptic failure precedes the loss of neurons and decline in cognitive function in neurodegenerative disorders, but the specific proteins and posttranslational modifications that promote synaptic failure in vascular dementia (VaD) remain largely unknown. We therefore used an isobaric tag for relative and absolute proteomic quantitation (iTRAQ) to profile the synapse-associated proteome of post-mortem human cortex from vascular dementia patients and age-matched controls. Brain tissue from VaD patients exhibited significant down-regulation of critical synaptic proteins including clathrin (0.29; p < 1.0⋅10(-3)) and GDI1 (0.51; p = 3.0⋅10(-3)), whereas SNAP25 (1.6; p = 5.5⋅10(-3)), bassoon (1.4; p = 1.3⋅10(-3)), excitatory amino acid transporter 2 (2.6; p = 9.2⋅10(-3)) and Ca(2+)/calmodulin dependent kinase II (1.6; p = 3.0⋅10(-2)) were substantially up-regulated. Our analyses further revealed divergent patterns of protein modification in the dementia patient samples, including a specific deamidation of synapsin1 predicted to compromise protein structure. Our results reveal potential molecular targets for intervention in synaptic failure and prevention of cognitive decline in VaD.
Collapse
Affiliation(s)
| | - Aida Serra
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Jingru Qian
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Christopher P Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Memory, Aging and Cognition Centre, National University Health System, Singapore
| | - Raj N Kalaria
- Institute for Ageing and Health, NIHR Biomedical Research Building, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, United Kingdom
| | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University, Singapore.
| |
Collapse
|
36
|
Weber JP, Toft-Bertelsen TL, Mohrmann R, Delgado-Martinez I, Sørensen JB. Synaptotagmin-7 is an asynchronous calcium sensor for synaptic transmission in neurons expressing SNAP-23. PLoS One 2014; 9:e114033. [PMID: 25422940 PMCID: PMC4244210 DOI: 10.1371/journal.pone.0114033] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 11/03/2014] [Indexed: 12/25/2022] Open
Abstract
Synchronization of neurotransmitter release with the presynaptic action potential is essential for maintaining fidelity of information transfer in the central nervous system. However, synchronous release is frequently accompanied by an asynchronous release component that builds up during repetitive stimulation, and can even play a dominant role in some synapses. Here, we show that substitution of SNAP-23 for SNAP-25 in mouse autaptic glutamatergic hippocampal neurons results in asynchronous release and a higher frequency of spontaneous release events (mEPSCs). Use of neurons from double-knock-out (SNAP-25, synaptotagmin-7) mice in combination with viral transduction showed that SNAP-23-driven release is triggered by endogenous synaptotagmin-7. In the absence of synaptotagmin-7 release became even more asynchronous, and the spontaneous release rate increased even more, indicating that synaptotagmin-7 acts to synchronize release and suppress spontaneous release. However, compared to synaptotagmin-1, synaptotagmin-7 is a both leaky and asynchronous calcium sensor. In the presence of SNAP-25, consequences of the elimination of synaptotagmin-7 were small or absent, indicating that the protein pairs SNAP-25/synaptotagmin-1 and SNAP-23/synaptotagmin-7 might act as mutually exclusive calcium sensors. Expression of fusion proteins between pHluorin (pH-sensitive GFP) and synaptotagmin-1 or -7 showed that vesicles that fuse using the SNAP-23/synaptotagmin-7 combination contained synaptotagmin-1, while synaptotagmin-7 barely displayed activity-dependent trafficking between vesicle and plasma membrane, implying that it acts as a plasma membrane calcium sensor. Overall, these findings support the idea of alternative syt∶SNARE combinations driving release with different kinetics and fidelity.
Collapse
Affiliation(s)
- Jens P. Weber
- Department of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, The Netherlands
| | - Trine L. Toft-Bertelsen
- Department of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ralf Mohrmann
- Department of Physiology, University of Saarland, Homburg, Germany
| | | | - Jakob B. Sørensen
- Department of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Lundbeck Foundation Center for Biomembranes in Nanomedicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
37
|
Ramirez DMO, Kavalali ET. The role of non-canonical SNAREs in synaptic vesicle recycling. CELLULAR LOGISTICS 2014; 2:20-27. [PMID: 22645707 PMCID: PMC3355972 DOI: 10.4161/cl.20114] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
An increasing number of studies suggest that distinct pools of synaptic vesicles drive specific forms of neurotransmission. Interspersed with these functional studies are analyses of the synaptic vesicle proteome which have consistently detected the presence of so-called “non-canonical” SNAREs that typically function in fusion and trafficking of other subcellular structures within the neuron. The recent identification of certain non-canonical vesicular SNAREs driving spontaneous (e.g., VAMP7 and vti1a) or evoked asynchronous (e.g., VAMP4) release integrates and corroborates existing data from functional and proteomic studies and implies that at least some complement of non-canonical SNAREs resident on synaptic vesicles function in neurotransmission. Here, we discuss the specific roles in neurotransmission of proteins homologous to each member of the classical neuronal SNARE complex consisting of synaptobrevin2, syntaxin-1 and SNAP-25.
Collapse
|
38
|
Horváth E, Woodhams SG, Nyilas R, Henstridge CM, Kano M, Sakimura K, Watanabe M, Katona I. Heterogeneous presynaptic distribution of monoacylglycerol lipase, a multipotent regulator of nociceptive circuits in the mouse spinal cord. Eur J Neurosci 2014; 39:419-34. [PMID: 24494682 PMCID: PMC3979158 DOI: 10.1111/ejn.12470] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 11/29/2013] [Accepted: 12/02/2013] [Indexed: 01/10/2023]
Abstract
Monoacylglycerol lipase (MGL) is a multifunctional serine hydrolase, which terminates anti-nociceptive endocannabinoid signaling and promotes pro-nociceptive prostaglandin signaling. Accordingly, both acute nociception and its sensitization in chronic pain models are prevented by systemic or focal spinal inhibition of MGL activity. Despite its analgesic potential, the neurobiological substrates of beneficial MGL blockade have remained unexplored. Therefore, we examined the regional, cellular and subcellular distribution of MGL in spinal circuits involved in nociceptive processing. All immunohistochemical findings obtained with light, confocal or electron microscopy were validated in MGL-knockout mice. Immunoperoxidase staining revealed a highly concentrated accumulation of MGL in the dorsal horn, especially in superficial layers. Further electron microscopic analysis uncovered that the majority of MGL-immunolabeling is found in axon terminals forming either asymmetric glutamatergic or symmetric γ-aminobutyric acid/glycinergic synapses in laminae I/IIo. In line with this presynaptic localization, analysis of double-immunofluorescence staining by confocal microscopy showed that MGL colocalizes with neurochemical markers of peptidergic and non-peptidergic nociceptive terminals, and also with markers of local excitatory or inhibitory interneurons. Interestingly, the ratio of MGL-immunolabeling was highest in calcitonin gene-related peptide-positive peptidergic primary afferents, and the staining intensity of nociceptive terminals was significantly reduced in MGL-knockout mice. These observations highlight the spinal nociceptor synapse as a potential anatomical site for the analgesic effects of MGL blockade. Moreover, the presence of MGL in additional terminal types raises the possibility that MGL may play distinct regulatory roles in synaptic endocannabinoid or prostaglandin signaling according to its different cellular locations in the dorsal horn pain circuitry.
Collapse
Affiliation(s)
- Eszter Horváth
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony utca 43., H-1083, Budapest, Hungary
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Meriney SD, Umbach JA, Gundersen CB. Fast, Ca2+-dependent exocytosis at nerve terminals: shortcomings of SNARE-based models. Prog Neurobiol 2014; 121:55-90. [PMID: 25042638 DOI: 10.1016/j.pneurobio.2014.07.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 04/14/2014] [Accepted: 07/03/2014] [Indexed: 11/30/2022]
Abstract
Investigations over the last two decades have made major inroads in clarifying the cellular and molecular events that underlie the fast, synchronous release of neurotransmitter at nerve endings. Thus, appreciable progress has been made in establishing the structural features and biophysical properties of the calcium (Ca2+) channels that mediate the entry into nerve endings of the Ca2+ ions that trigger neurotransmitter release. It is now clear that presynaptic Ca2+ channels are regulated at many levels and the interplay of these regulatory mechanisms is just beginning to be understood. At the same time, many lines of research have converged on the conclusion that members of the synaptotagmin family serve as the primary Ca2+ sensors for the action potential-dependent release of neurotransmitter. This identification of synaptotagmins as the proteins which bind Ca2+ and initiate the exocytotic fusion of synaptic vesicles with the plasma membrane has spurred widespread efforts to reveal molecular details of synaptotagmin's action. Currently, most models propose that synaptotagmin interfaces directly or indirectly with SNARE (soluble, N-ethylmaleimide sensitive factor attachment receptors) proteins to trigger membrane fusion. However, in spite of intensive efforts, the field has not achieved consensus on the mechanism by which synaptotagmins act. Concurrently, the precise sequence of steps underlying SNARE-dependent membrane fusion remains controversial. This review considers the pros and cons of the different models of SNARE-mediated membrane fusion and concludes by discussing a novel proposal in which synaptotagmins might directly elicit membrane fusion without the intervention of SNARE proteins in this final fusion step.
Collapse
Affiliation(s)
- Stephen D Meriney
- Department of Neuroscience, Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Joy A Umbach
- Department of Molecular and Medical Pharmacology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Cameron B Gundersen
- Department of Molecular and Medical Pharmacology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA.
| |
Collapse
|
40
|
Vecchia D, Tottene A, van den Maagdenberg AMJM, Pietrobon D. Mechanism underlying unaltered cortical inhibitory synaptic transmission in contrast with enhanced excitatory transmission in CaV2.1 knockin migraine mice. Neurobiol Dis 2014; 69:225-34. [PMID: 24907493 PMCID: PMC4107271 DOI: 10.1016/j.nbd.2014.05.035] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 05/07/2014] [Accepted: 05/27/2014] [Indexed: 02/03/2023] Open
Abstract
Familial hemiplegic migraine type 1 (FHM1), a monogenic subtype of migraine with aura, is caused by gain-of-function mutations in CaV2.1 (P/Q-type) calcium channels. In FHM1 knockin mice, excitatory neurotransmission at cortical pyramidal cell synapses is enhanced, but inhibitory neurotransmission at connected pairs of fast-spiking (FS) interneurons and pyramidal cells is unaltered, despite being initiated by CaV2.1 channels. The mechanism underlying the unaltered GABA release at cortical FS interneuron synapses remains unknown. Here, we show that the FHM1 R192Q mutation does not affect inhibitory transmission at autapses of cortical FS and other types of multipolar interneurons in microculture from R192Q knockin mice, and investigate the underlying mechanism. Lowering the extracellular [Ca2+] did not reveal gain-of-function of evoked transmission neither in control nor after prolongation of the action potential (AP) with tetraethylammonium, indicating unaltered AP-evoked presynaptic calcium influx at inhibitory autapses in FHM1 KI mice. Neither saturation of the presynaptic calcium sensor nor short duration of the AP can explain the unaltered inhibitory transmission in the mutant mice. Recordings of the P/Q-type calcium current in multipolar interneurons in microculture revealed that the current density and the gating properties of the CaV2.1 channels expressed in these interneurons are barely affected by the FHM1 mutation, in contrast with the enhanced current density and left-shifted activation gating of mutant CaV2.1 channels in cortical pyramidal cells. Our findings suggest that expression of specific CaV2.1 channels differentially sensitive to modulation by FHM1 mutations in inhibitory and excitatory cortical neurons underlies the gain-of-function of excitatory but unaltered inhibitory synaptic transmission and the likely consequent dysregulation of the cortical excitatory–inhibitory balance in FHM1. Multipolar interneuron autaptic transmission is unaltered in FHM1 knockin mice. This is due to unaltered action potential (AP)-evoked presynaptic Ca influx. Unaltered AP-evoked Ca influx is not due to short duration of interneuron APs. CaV2.1 channels of multipolar interneurons are barely affected by the FHM1 mutation. This may explain unaltered inhibitory neurotransmission in FHM1.
Collapse
Affiliation(s)
- Dania Vecchia
- Department of Biomedical Sciences, University of Padova and CNR Institute of Neuroscience, 35121 Padova, Italy
| | - Angelita Tottene
- Department of Biomedical Sciences, University of Padova and CNR Institute of Neuroscience, 35121 Padova, Italy
| | | | - Daniela Pietrobon
- Department of Biomedical Sciences, University of Padova and CNR Institute of Neuroscience, 35121 Padova, Italy.
| |
Collapse
|
41
|
Falco A, Pennucci R, Brambilla E, de Curtis I. Reduction in parvalbumin-positive interneurons and inhibitory input in the cortex of mice with experimental autoimmune encephalomyelitis. Exp Brain Res 2014; 232:2439-49. [PMID: 24770856 PMCID: PMC4055863 DOI: 10.1007/s00221-014-3944-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 04/02/2014] [Indexed: 01/30/2023]
Abstract
In multiple sclerosis (MS), inflammation leads to damage of central nervous system myelin and axons. Previous studies have postulated impaired GABA transmission in MS, and recent postmortem analysis has shown that GABAergic parvalbumin (PV)-positive interneurons are decreased in the primary motor cortex (M1) of patients with MS. In this report, we present evidence for the loss of a specific population of GABAergic interneurons in the experimental autoimmune encephalomyelitis mouse model of MS. Using experimental autoimmune encephalomyelitis, we evaluated the distribution of both PV-positive interneurons and of the inhibitory presynaptic input in the M1 of experimental autoimmune encephalomyelitis and control mice. Our results demonstrate a specific decrease in the number of PV-positive interneurons in the M1 of mice with experimental autoimmune encephalomyelitis. We detected a significant reduction in the number of PV-positive interneurons in the layers II and III of the M1 of diseased mice, while there was no difference in the number of calretinin (CR)-positive cells between animals with experimental autoimmune encephalomyelitis and control animals. Moreover, we observed a significant reduction in the inhibitory presynaptic input in the M1 of treated mice. These changes were specific for the mice with elevated clinical score, while they were not detectable in the mice with low clinical score. Our results support the hypothesis that reinforcing the action of the GABAergic network may represent a therapeutic alternative to limit the progression of the neuronal damage in MS patients.
Collapse
Affiliation(s)
- Anna Falco
- Cell Adhesion Unit, Division of Neuroscience, San Raffaele Scientific Institute, via Olgettina, 58-20132, Milan, Italy
| | | | | | | |
Collapse
|
42
|
Distribution of SNAP25, VAMP1 and VAMP2 in mature and developing deep cerebellar nuclei after estrogen administration. Neuroscience 2014; 266:102-15. [DOI: 10.1016/j.neuroscience.2014.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 02/04/2014] [Accepted: 02/06/2014] [Indexed: 11/19/2022]
|
43
|
Yilmaz M, Edgunlu TG, Yilmaz N, Cetin ES, Celik SK, Emir GK, Sözen A. Genetic variants of synaptic vesicle and presynaptic plasma membrane proteins in idiopathic generalized epilepsy. J Recept Signal Transduct Res 2013; 34:38-43. [PMID: 24164654 DOI: 10.3109/10799893.2013.848893] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND The aim of this study was to analyze the role of the genetic variants of two synaptic vesicle proteins (VAMP2 and Synaptotagmin XI) and two presynaptic plasma membrane proteins (Syntaxin 1A and SNAP-25) in patients with idiopathic generalized epilepsy (IGE). METHOD Eighty-five patients with IGE and 93 healthy subjects were included in the study. We analyzed the functional polymorphisms of VAMP2, Synaptotagmin XI, Syntaxin 1A and SNAP-25 genes with polymerase chain reaction and restriction fragment length polymorphism methods. RESULTS In the patients with IGE, significant differences alleles and genotypes of 26 bp Ins/Del polymorphism of the VAMP2 gene and the 33-bp promoter region of Synaptotagmin XI were observed, however no associaton was found regarding Intron 7 rs1569061 of Syntaxin 1A gene, MnlI rs3746544 and DdeI rs1051312 polymorphisms of SNAP-25 gene compared with healthy subjects. Carriers of the C allele of Synaptotagmin XI had worse measures compared with the T allele of Synaptotagmin XI. In the haplotype analysis, the frequency of the T alleles of rs1569061 and of the C alleles of the 33-bp promoter region of Synaptotagmin XI was found to be significantly higher in patients with IGE as compared with the healthy subjects. CONCLUSION The genetic variations of VAMP2, Synaptotagmin XI might be indication of the relationship between these genes and IGE.
Collapse
|
44
|
Peng L, Liu H, Ruan H, Tepp WH, Stoothoff WH, Brown RH, Johnson EA, Yao WD, Zhang SC, Dong M. Cytotoxicity of botulinum neurotoxins reveals a direct role of syntaxin 1 and SNAP-25 in neuron survival. Nat Commun 2013; 4:1472. [PMID: 23403573 PMCID: PMC4052923 DOI: 10.1038/ncomms2462] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 01/09/2013] [Indexed: 12/05/2022] Open
Abstract
Botulinum neurotoxins (BoNT/A-G) are well-known to act by blocking synaptic vesicle exocytosis. Whether BoNTs disrupt additional neuronal functions has not been addressed. Here we report that cleavage of syntaxin 1 (Syx 1) by BoNT/C and cleavage of SNAP-25 by BoNT/E both induce degeneration of cultured rodent and human neurons. Furthermore, although SNAP-25 cleaved by BoNT/A can still support neuron survival, it has reduced capacity to tolerate additional mutations and also fails to pair with syntaxin isoforms other than Syx 1. Syx 1 and SNAP-25 are well-known for mediating synaptic vesicle exocytosis, but we found that neuronal death is due to blockage of plasma membrane recycling processes that share Syx 1/SNAP-25 for exocytosis, independent of blockage of synaptic vesicle exocytosis. These findings reveal neuronal cytotoxicity for a subset of BoNTs and directly link Syx 1/SNAP-25 to neuron survival as the prevalent SNARE proteins mediating multiple fusion events on neuronal plasma membranes.
Collapse
Affiliation(s)
- Lisheng Peng
- Department of Microbiology and Immunobiology, Harvard Medical School, Division of Neuroscience, New England Primate Research Center, Southborough, Massachusetts 01772, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Reduced SNAP-25 alters short-term plasticity at developing glutamatergic synapses. EMBO Rep 2013; 14:645-51. [PMID: 23732542 DOI: 10.1038/embor.2013.75] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 05/13/2013] [Accepted: 05/14/2013] [Indexed: 02/07/2023] Open
Abstract
SNAP-25 is a key component of the synaptic-vesicle fusion machinery, involved in several psychiatric diseases including schizophrenia and ADHD. SNAP-25 protein expression is lower in different brain areas of schizophrenic patients and in ADHD mouse models. How the reduced expression of SNAP-25 alters the properties of synaptic transmission, leading to a pathological phenotype, is unknown. We show that, unexpectedly, halved SNAP-25 levels at 13-14 DIV not only fail to impair synaptic transmission but instead enhance evoked glutamatergic neurotransmission. This effect is possibly dependent on presynaptic voltage-gated calcium channel activity and is not accompanied by changes in spontaneous quantal events or in the pool of readily releasable synaptic vesicles. Notably, synapses of 13-14 DIV neurons with reduced SNAP-25 expression show paired-pulse depression as opposed to paired-pulse facilitation occurring in their wild-type counterparts. This phenotype disappears with synapse maturation. As alterations in short-term plasticity represent a new mechanism contributing to cognitive impairments in intellectual disabilities, our data provide mechanistic clues for neuronal circuit alterations in psychiatric diseases characterized by reduced expression of SNAP-25.
Collapse
|
46
|
Proteomic analysis of adrenocorticotropic hormone treatment of an infantile spasm model induced by N-methyl-D-aspartic acid and prenatal stress. PLoS One 2012; 7:e45347. [PMID: 23028951 PMCID: PMC3445469 DOI: 10.1371/journal.pone.0045347] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 08/21/2012] [Indexed: 11/19/2022] Open
Abstract
Infantile spasms is an age-specific epileptic syndrome associated with poor developmental outcomes and poor response to nearly all traditional antiepileptic drugs except adrenocorticotropic hormone (ACTH). We investigated the protective mechanism of ACTH against brain damage. An infantile spasm rat model induced by N-methyl-d-aspartate (NMDA) in neonate rats was used. Pregnant rats were randomly divided into the stress-exposed and the non-stress exposed groups, and their offspring were randomly divided into ACTH-treated spasm model, untreated spasm model, and control groups. A proteomics-based approach was used to detect the proteome differences between ACTH-treated and untreated groups. Gel image analysis was followed by matrix-assisted laser desorption/ionization time-of-flight mass spectrometric protein identification and bioinformatics analysis. Prenatal stress exposure resulted in more severe seizures, and ACTH treatment reduced and delayed the onset of seizures. The most significantly up-regulated proteins included isoform 1 of tubulin β-5 chain, cofilin-1 (CFL1), synaptosomal-associated protein 25, malate dehydrogenase, N(G),N(G)-dimethylarginine dimethylaminohydrolase 1, annexin A3 (ANXA3), and rho GDP-dissociation inhibitor 1 (ARHGDIA). In contrast, tubulin α-1A chain was down-regulated. Three of the identified proteins, ARHGDIA, ANXA3, and CFL1, were validated using western blot analysis. ARHGDIA expression was assayed in the brain samples of five infantile spasm patients. These proteins are involved in the cytoskeleton, synapses, energy metabolism, vascular regulation, signal transduction, and acetylation. The mechanism underlying the effects of ACTH involves the molecular events affected by these proteins, and protein acetylation is the mechanism of action of the drug treatment.
Collapse
|
47
|
Blakey D, Wilson MC, Molnár Z. Termination and initial branch formation of SNAP-25-deficient thalamocortical fibres in heterochronic organotypic co-cultures. Eur J Neurosci 2012; 35:1586-94. [PMID: 22607004 PMCID: PMC3359864 DOI: 10.1111/j.1460-9568.2012.08120.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
We are interested in the role of neural activity mediated through regulated vesicular release in the stopping and early branching of the thalamic projections in the cortex. Axon outgrowth, arrival at the cortical subplate, side-branch formation during the waiting period and cortical plate innervation of embryonic thalamocortical projections occurs without major abnormalities in the absence of regulated release in Snap25 (-/-) null mutant mice [Washbourne et al. (2002) Nat. Neurosci. 5:19-26; Molnár et al. (2002) J. Neurosci. 22:10313-10323]. The fact that Snap25 (-/-) null mutant mice die at birth limited our previous experiments to the prenatal period. We therefore investigated the behaviour of thalamic projections in co-culture paradigms by using heterochronic thalamic [embryonic day (E)16-E18] and cortical [postnatal day (P)0-P3] explants, in which the stopping and branching behaviour has been previously documented. Our current co-culture experiments established that thalamic projections from E16-E18 Snap25(+/+) or Snap25 (-/-) explants behaved in an identical fashion in P0-P3 Snap25 (+/+) cortical explants after 7 days in vitro. Thalamic projections from Snap25 (-/-) explants developed similar patterns of fibre ingrowth to the cortex, and stopped and formed branches at a similar depth in the Snap25(+/+) cortical slice as in control cultures. These results imply that thalamic projections can reach their ultimate target cells in layer 4, stop, and start to develop branches in the absence of regulated vesicular transmitter release from their own terminals.
Collapse
Affiliation(s)
- Daniel Blakey
- Department of Physiology, Anatomy and Genetics, Le Gros Clark Building, University of Oxford, South Parks Road, Oxford OX1 3QX
| | - Michael C Wilson
- Department of Neuroscience, University of New Mexico MSC08 4740 1, Albuquerque NM 87131-5223, USA
| | - Zoltán Molnár
- Department of Physiology, Anatomy and Genetics, Le Gros Clark Building, University of Oxford, South Parks Road, Oxford OX1 3QX
| |
Collapse
|
48
|
Rowley NM, Madsen KK, Schousboe A, Steve White H. Glutamate and GABA synthesis, release, transport and metabolism as targets for seizure control. Neurochem Int 2012; 61:546-58. [PMID: 22365921 DOI: 10.1016/j.neuint.2012.02.013] [Citation(s) in RCA: 122] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 02/09/2012] [Indexed: 11/17/2022]
Abstract
The synthesis, release, reuptake, and metabolism of the excitatory and inhibitory neurotransmitters glutamate and GABA, respectively, are tightly controlled. Given the role that these two neurotransmitters play in normal and abnormal neurotransmission, it is important to consider the processes whereby they are regulated. This brief review is focused entirely on the metabolic aspects of glutamate and GABA synthesis and neurotransmission. It describes in limited detail the synthesis, release, reuptake, metabolism, cellular compartmentation and pharmacology of the glutamatergic and GABAergic synapse. This review also provides a summary and brief description of the pathologic and phenotypic features of the various genetic animal models that have been developed in an effort to provide a greater understanding of the role that each of the aforementioned metabolic processes plays in controlling excitatory and inhibitory neurotransmission and how their use will hopefully facilitate the development of safer and more efficacious therapies for the treatment of epilepsy and other neurological disorders.
Collapse
Affiliation(s)
- Nicole M Rowley
- Department of Pharmacology and Toxicology, Anticonvulsant Drug Development Program, University of Utah, 417 Wakara Way, Suite 3211, Salt Lake City, UT 84108, USA
| | | | | | | |
Collapse
|
49
|
Bragina L, Fattorini G, Giovedí S, Melone M, Bosco F, Benfenati F, Conti F. Analysis of Synaptotagmin, SV2, and Rab3 Expression in Cortical Glutamatergic and GABAergic Axon Terminals. Front Cell Neurosci 2012; 5:32. [PMID: 22275882 PMCID: PMC3254050 DOI: 10.3389/fncel.2011.00032] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Accepted: 12/25/2011] [Indexed: 11/13/2022] Open
Abstract
We investigated whether cortical glutamatergic and GABAergic release machineries can be differentiated on the basis of the nature and amount of proteins they express, by performing a quantitative analysis of the degree of co-localization of synaptotagmin (SYT) 1 and 2, synaptic vesicle protein 2 (SV2) A and B, and Rab3a and c in VGLUT1+, VGLUT2+, and VGAT+ terminals and synaptic vesicles (SVs) in rat cerebral cortex. Co-localization studies showed that VGLUT1 puncta had high levels of SV2A and B and of Rab3c, intermediate levels of SYT1, and low levels of SYT2 and Rab3c; VGLUT2 puncta exhibited intermediate levels of all presynaptic proteins studied; whereas vesicular GABA transporter (VGAT) puncta had high levels of SV2A and SYT2, intermediate levels of SYT1, Rab3a, and Rab3c, and low levels of SV2B. Since SV2B is reportedly expressed by glutamatergic neurons and we observed SV2B expression in VGAT puncta, we performed electron microscopic studies and found SV2B positive axon terminals forming symmetric synapses. Immunoisolation studies showed that the expression levels of the protein isoforms varied in the three populations of SVs. Expression of SYT1 was highest in VGLUT1–SVs, while SYT2 expression was similar in the three SV groups. Expression of SV2A was similarly high in all three SV populations, except for SV2B levels that were very low in VGAT SVs. Finally, Rab3a levels were similar in the three SV groups, while Rab3c levels were highest in VGLUT1–SVs. These quantitative results extend our previous studies on the differential expression of presynaptic proteins involved in neurotransmitter release in GABAergic and glutamatergic terminals and indicate that heterogeneity of the respective release machineries can be generated by the differential complement of SV proteins involved in distinct stages of the release process.
Collapse
Affiliation(s)
- Luca Bragina
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche Ancona, Italy
| | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Dendritic exocytosis underpins a broad range of integrative and homeostatic synaptic functions. Emerging data highlight the essential role of SNAREs in trafficking and fusion of secretory organelles with release of peptides and neurotransmitters from dendrites. This Perspective analyzes recent evidence inferring axo-dendritic polarization of vesicular release machinery and pinpoints progress made with existing challenges in this rapidly progressing field of dendritic research. Interpreting the relation of new molecular data to physiological results on secretion from dendrites would greatly advance our understanding of this facet of neuronal mechanisms.
Collapse
Affiliation(s)
- Saak V. Ovsepian
- International Centre for Neurotherapeutics, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - J. Oliver Dolly
- International Centre for Neurotherapeutics, Dublin City University, Glasnevin, Dublin 9, Ireland
| |
Collapse
|