1
|
Ji H, Kim KR, Park JJ, Lee JY, Sim Y, Choi H, Kim S. Combination Gene Delivery Reduces Spinal Cord Pathology in Rats With Peripheral Neuropathic Pain. THE JOURNAL OF PAIN 2023; 24:2211-2227. [PMID: 37442406 DOI: 10.1016/j.jpain.2023.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 06/25/2023] [Accepted: 07/06/2023] [Indexed: 07/15/2023]
Abstract
Although peripheral neuropathic pain is caused by peripheral nerve injury, it is not simply a peripheral nervous system disease. It causes abnormalities in both the central and peripheral nervous systems. Pathological phenomena, such as hyperactivation of sensory neurons and inflammation, are observed in both the dorsal root ganglion and spinal cord. Pain signals originating from the periphery are transmitted to the brain via the SC, and the signals are modulated by pathologically changing SC conditions. Therefore, the modulation of SC pathology is important for peripheral NP treatment. We investigated the effects of KLS-2031 (recombinant adeno-associated viruses expressing glutamate decarboxylase 65, glial cell-derived neurotrophic factor, and interleukin-10) delivered to the dorsal root ganglion on aberrant neuronal excitability and neuroinflammation in the SC of rats with peripheral NP. Results showed that KLS-2031 administration restored excessive excitatory transmission and inhibitory signals in substantia gelatinosa neurons. Moreover, KLS-2031 restored the in vivo hypersensitivity of wide dynamic range neurons and mitigated neuroinflammation in the SC by regulating microglia and astrocytes. Collectively, these findings demonstrated that KLS-2031 efficiently suppressed pathological pain signals and inflammation in the SC of peripheral NP model, and is a potential novel therapeutic approach for NP in clinical settings. PERSPECTIVE: Our study demonstrated that KLS-2031, a combination gene therapy delivered by transforaminal epidural injection, not only mitigates neuroinflammation but also improves SC neurophysiological function, including excitatory-inhibitory balance. These findings support the potential of KLS-2031 as a novel modality that targets multiple aspects of the complex pathophysiology of neuropathic pain.
Collapse
Affiliation(s)
- Hyelin Ji
- Institute of BioInnovation Research, Kolon Life Science, Seoul, Republic of Korea
| | - Kyung-Ran Kim
- Institute of BioInnovation Research, Kolon Life Science, Seoul, Republic of Korea
| | - Jang-Joon Park
- Institute of BioInnovation Research, Kolon Life Science, Seoul, Republic of Korea
| | - Ju Youn Lee
- Institute of BioInnovation Research, Kolon Life Science, Seoul, Republic of Korea
| | - Yeomoon Sim
- Institute of BioInnovation Research, Kolon Life Science, Seoul, Republic of Korea; Business Development, Handok Inc., Seoul, Republic of Korea
| | - Heonsik Choi
- Institute of BioInnovation Research, Kolon Life Science, Seoul, Republic of Korea; Healthcare Research Institute, Kolon Advanced Research Center, Kolon Industries, Seoul, Republic of Korea
| | - Sujeong Kim
- Institute of BioInnovation Research, Kolon Life Science, Seoul, Republic of Korea
| |
Collapse
|
2
|
Esmaeili-Mahani S, Haghparast E, Nezhadi A, Abbasnejad M, Sheibani V. Apelin-13 prevents hippocampal synaptic plasticity impairment in Parkinsonism rats. J Chem Neuroanat 2020; 111:101884. [PMID: 33161074 DOI: 10.1016/j.jchemneu.2020.101884] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/13/2022]
Abstract
The hippocampus is involved in learning and memory for novel information and implicated within the cognitive dysfunction in Parkinson's disease. Long-term potentiation (LTP), the most type of synaptic plasticity, is the base of learning and memory. We evaluated the consequences of apelin-13 on early long-term potentiation (E-LTP) in the Cornu Ammonis (CA1) area of the hippocampus and synaptic hippocampal protein expression of postsynaptic density protein 95 (PSD-95) and dopaminergic receptor (DR1) of the rat model of Parkinsonism. 6-hydroxydopamine (6-OHDA) was infused within the right substantia nigra. Intra-nigral transfusion of apelin-13 (1, 2, and 3 μg/rat) was performed one week after the 6-OHDA injection. Using hematoxylin and eosin staining, the pathological changes in the substantia nigra neurons were examined. In Vivo field excitatory postsynaptic potentials were recorded in the CA1 region one month after the apelin injection. The PSD-95 and DR1 protein levels were assessed by western blotting. The mRNA expression level of DR1 was also measured by real-time PCR. 6-OHDA meaningfully disrupted short-term memory and LTP, and altered the expression levels of the above-mentioned proteins in the hippocampus. The results suggest that apelin-13 (especially at 3 μg/rat) significantly ameliorates the E-LTP impairment and attenuates the changes in hippocampal synaptic proteins in 6-OHDA-treated rats.
Collapse
Affiliation(s)
- Saeed Esmaeili-Mahani
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran; Laboratory of Molecular Neuroscience, Kerman Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Elham Haghparast
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran; Laboratory of Molecular Neuroscience, Kerman Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | - Akram Nezhadi
- Neuroscience Research Center, Aja University of Medical Sciences, Tehran, Iran
| | - Mehdi Abbasnejad
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Vahid Sheibani
- Laboratory of Molecular Neuroscience, Kerman Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
3
|
Ma YC, Yang B, Wang X, Zhou L, Li WY, Liu WS, Lu XH, Zheng ZH, Ma Y, Wang RL. Identification of novel inhibitor of protein tyrosine phosphatases delta: structure-based pharmacophore modeling, virtual screening, flexible docking, molecular dynamics simulation, and post-molecular dynamics analysis. J Biomol Struct Dyn 2019; 38:4432-4448. [PMID: 31625456 DOI: 10.1080/07391102.2019.1682050] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Owing to their unique functions in regulating the synapse activity of protein tyrosine phosphatases delta (PTPδ) that has drawn special attention for developing drugs to autism spectrum disorders (ASDs). In this study, the PTPδ pharmacophore was first established by the structure-based pharmacophore method. Subsequently, 10 compounds contented Lipinski's rule of five was acquired by the virtual screening of the PTPδ pharmacophore against ZINC and PubChem databases. Then, the 10 identified molecules were discovered that had better binding affinity than a known PTPδ inhibitors compound SCHEMBL16375396. Two compounds SCHEMBL16375408 and ZINC19796658 with high binding score, low toxicity were gained. They were observed by docking analysis and molecular dynamics simulations that the novel potential inhibitors not only possessed the same function as SCHEMBL16375396 did in inhibiting PTPδ, but also had more favorable conformation to bind with the catalytic active regions. This study provides a new method for identify PTPδ inhibitor for the treatment of ASDs disease.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Yang-Chun Ma
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Bing Yang
- Department of Cell Biology, School of Basic Medical Science, Tianjin Medical University, Tianjin, China
| | - Xin Wang
- Tasly Pharmaceutical Group Co., Ltd., Tianjin, China
| | - Liang Zhou
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Wei-Ya Li
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Wen-Shan Liu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Xin-Hua Lu
- New Drug Research and Development Center of North China Pharmaceutical Group Corporation, National Microbial Medicine Engineering and Research Center, Hebei Industry Microbial Metabolic Engineering & Technology Research Center, Key Laboratory for New Drug Screening Technology of Shijiazhuang City, Shijiazhuang, Hebei, China
| | - Zhi-Hui Zheng
- New Drug Research and Development Center of North China Pharmaceutical Group Corporation, National Microbial Medicine Engineering and Research Center, Hebei Industry Microbial Metabolic Engineering & Technology Research Center, Key Laboratory for New Drug Screening Technology of Shijiazhuang City, Shijiazhuang, Hebei, China
| | - Ying Ma
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Run-Ling Wang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| |
Collapse
|
4
|
Morales-Ovalles Y, Miranda-Contreras L, Peña-Contreras Z, Dávila-Vera D, Balza-Quintero A, Sánchez-Gil B, Mendoza-Briceño RV. Developmental exposure to mancozeb induced neurochemical and morphological alterations in adult male mouse hypothalamus. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2018; 64:139-146. [PMID: 30391875 DOI: 10.1016/j.etap.2018.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 10/11/2018] [Accepted: 10/16/2018] [Indexed: 06/08/2023]
Abstract
Mancozeb, a dithiocarbamate widely used in agriculture, is considered a developmental hazard in humans; however, more evidences are still needed concerning the consequences of chronic exposure to this pesticide. Mancozeb neurotoxicity in developing mouse hypothalamus was evaluated by subchronic exposure of male Mus musculus mice to low and high doses of mancozeb (30 and 90 mg/kg body weight, respectively) from late neonatal until adolescence. Variations in hypothalamic amino acid neurotransmitter levels and changes in histological as well as cytological characteristics were analyzed in young adult experimental mice and compared with control. A dose-dependent increase in excitation/ inhibition ratio was observed in mancozeb-exposed hypothalamus, indicating an overall state of excitoxicity. Histopathological and ultrastructural studies showed increased apoptosis, neuroinflammation and demyelination, demonstrating mancozeb-induced cytotoxicity in hypothalamic neurosecretory cells. In summary, both neurochemical and morphological data revealed mancozeb-induced alterations during development of hypothalamic circuitry that are critical for maturation of the neuroendocrine system.
Collapse
Affiliation(s)
- Yasmin Morales-Ovalles
- Electron Microscopy Center "Dr. Ernesto Palacios Prü", University of Los Andes, Mérida, Venezuela
| | | | - Zulma Peña-Contreras
- Electron Microscopy Center "Dr. Ernesto Palacios Prü", University of Los Andes, Mérida, Venezuela
| | - Delsy Dávila-Vera
- Electron Microscopy Center "Dr. Ernesto Palacios Prü", University of Los Andes, Mérida, Venezuela
| | - Alirio Balza-Quintero
- Electron Microscopy Center "Dr. Ernesto Palacios Prü", University of Los Andes, Mérida, Venezuela
| | - Beluardi Sánchez-Gil
- Electron Microscopy Center "Dr. Ernesto Palacios Prü", University of Los Andes, Mérida, Venezuela
| | | |
Collapse
|
5
|
Jaramillo TC, Escamilla CO, Liu S, Peca L, Birnbaum SG, Powell CM. Genetic background effects in Neuroligin-3 mutant mice: Minimal behavioral abnormalities on C57 background. Autism Res 2017; 11:234-244. [PMID: 29028156 DOI: 10.1002/aur.1857] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 08/09/2017] [Indexed: 11/11/2022]
Abstract
Neuroligin-3 (NLGN3) is a postsynaptic cell adhesion protein that interacts with presynaptic ligands including neurexin-1 (NRXN1) [Ichtchenko et al., Journal of Biological Chemistry, 271, 2676-2682, 1996]. Mice harboring a mutation in the NLGN3 gene (NL3R451C) mimicking a mutation found in two brothers with autism spectrum disorder (ASD) were previously generated and behaviorally phenotyped for autism-related behaviors. In these NL3R451C mice generated and tested on a hybrid C57BL6J/129S2/SvPasCrl background, we observed enhanced spatial memory and reduced social interaction [Tabuchi et al., Science, 318, 71-76, 2007]. Curiously, an independently generated second line of mice harboring the same mutation on a C57BL6J background exhibited minimal aberrant behavior, thereby providing apparently discrepant results. To investigate the origin of the discrepancy, we previously replicated the original findings of Tabuchi et al. by studying the same NL3R451C mutation on a pure 129S2/SvPasCrl genetic background. Here we complete the behavioral characterization of the NL3R451C mutation on a pure C57BL6J genetic background to determine if background genetics play a role in the discrepant behavioral outcomes involving NL3R451C mice. NL3R451C mutant mice on a pure C57BL6J background did not display spatial memory enhancements or social interaction deficits. We only observed a decreased startle response and mildly increased locomotor activity in these mice suggesting that background genetics influences behavioral outcomes involving the NL3R451C mutation. Autism Res 2018, 11: 234-244. © 2017 International Society for Autism Research, Wiley Periodicals, Inc. LAY SUMMARY Behavioral symptoms of autism can be highly variable, even in cases that involve identical genetic mutations. Previous studies in mice with a mutation of the Neuroligin-3 gene showed enhanced learning and social deficits. We replicated these findings on the same and different genetic backgrounds. In this study, however, the same mutation in mice on a different genetic background did not reproduce our previous findings. Our results suggest that genetic background influences behavioral symptoms of this autism-associated mutation.
Collapse
Affiliation(s)
- Thomas C Jaramillo
- Department of Neurology & Neurotherapeutics, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-8813
| | - Christine Ochoa Escamilla
- Department of Neurology & Neurotherapeutics, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-8813.,Neuroscience Graduate Program, The University of Texas Southwestern Medical Center, Dallas, TX, 75390
| | - Shunan Liu
- Department of Neurology & Neurotherapeutics, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-8813
| | - Lauren Peca
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, 75390-9170
| | - Shari G Birnbaum
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, 75390-9170
| | - Craig M Powell
- Department of Neurology & Neurotherapeutics, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-8813.,Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, 75390-9170.,Neuroscience Graduate Program, The University of Texas Southwestern Medical Center, Dallas, TX, 75390.,Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, TX, 75390
| |
Collapse
|
6
|
Cadherin-10 Maintains Excitatory/Inhibitory Ratio through Interactions with Synaptic Proteins. J Neurosci 2017; 37:11127-11139. [PMID: 29030434 DOI: 10.1523/jneurosci.1153-17.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 09/12/2017] [Accepted: 10/02/2017] [Indexed: 11/21/2022] Open
Abstract
Appropriate excitatory/inhibitory (E/I) balance is essential for normal cortical function and is altered in some psychiatric disorders, including autism spectrum disorders (ASDs). Cell-autonomous molecular mechanisms that control the balance of excitatory and inhibitory synapse function remain poorly understood; no proteins that regulate excitatory and inhibitory synapse strength in a coordinated reciprocal manner have been identified. Using super-resolution imaging, electrophysiology, and molecular manipulations, we show that cadherin-10, encoded by CDH10 within the ASD risk locus 5p14.1, maintains both excitatory and inhibitory synaptic scaffold structure in cultured cortical neurons from rats of both sexes. Cadherin-10 localizes to both excitatory and inhibitory synapses in neocortex, where it is organized into nanoscale puncta that influence the size of their associated PSDs. Knockdown of cadherin-10 reduces excitatory but increases inhibitory synapse size and strength, altering the E/I ratio in cortical neurons. Furthermore, cadherin-10 exhibits differential participation in complexes with PSD-95 and gephyrin, which may underlie its role in maintaining the E/I ratio. Our data provide a new mechanism whereby a protein encoded by a common ASD risk factor controls E/I ratios by regulating excitatory and inhibitory synapses in opposing directions.SIGNIFICANCE STATEMENT The correct balance between excitatory/inhibitory (E/I) is crucial for normal brain function and is altered in psychiatric disorders such as autism. However, the molecular mechanisms that underlie this balance remain elusive. To address this, we studied cadherin-10, an adhesion protein that is genetically linked to autism and understudied at the cellular level. Using a combination of advanced microscopy techniques and electrophysiology, we show that cadherin-10 forms nanoscale puncta at excitatory and inhibitory synapses, maintains excitatory and inhibitory synaptic structure, and is essential for maintaining the correct balance between excitation and inhibition in neuronal dendrites. These findings reveal a new mechanism by which E/I balance is controlled in neurons and may bear relevance to synaptic dysfunction in autism.
Collapse
|
7
|
Tian J, Li M, Zhao J, Li J, Liu G, Zhen Z, Cao Y, Gregersen H, Tong X. Research on the traditional Chinese medicine treating gastrointestinal motility in diabetic rats by improving biomechanical remodeling and neuroendocrine regulation. Am J Transl Res 2017; 9:2219-2230. [PMID: 28559973 PMCID: PMC5446505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 02/26/2017] [Indexed: 06/07/2023]
Abstract
Previous studies have demonstrated that TWA, a Chinese herbal medicine, could significantly improve the symptoms of patients with diabetic gastrointestinal dysfunction. However, the specific mechanism of regulating intestinal peristalsis has not been found. This study aimed to discover TWA's therapeutic mechanism for regulating intestinal motility. The intestinal propulsion rate of diabetic rats was significantly increased after treatment with TWA for 8 weeks. Aiming at the mechanical structure, biomechanical testing indicated that TWA can significantly decrease the no-load intestinal wall thickness, cross-sectional area, and angular spread in a zero-stress state. Notably, intestinal stress-strain curve shifted to the right, which indicated TWA can inhibit intestinal hyperplasia and hardening and improve biomechanical remodeling. Further study of the mechanism revealed that TWA significantly inhibited the expression of AGE in the villi, crypt, and muscle and RAGE in crypt and upregulated the expression of nerve regulator (PSD95, C-kit and SCF). Radioimmunoassay showed TWA treatment decreased levels of serum somatostatin and vasoactive intestinal peptide. Moreover, associations were found between the intestinal propulsion rate with the morphologic and biomechanical remodeling parameters, changes of nerve factors, and endocrine hormones. Morphologic and biomechanical remodeling of the intestinal wall are the pathologic basis of gastrointestinal dysfunction. TWA can benefit intestinal motility by improving biomechanical and morphologic remodeling and by regulating expression of neuroendocrine factors. The results showed that the effect of TWA was dose-dependent, the higher the dose, the greater is the improvement. Thus, traditional Chinese medicine might be a valuable tool for treating diabetic gastrointestinal dysfunction.
Collapse
Affiliation(s)
- Jiaxing Tian
- Graduate School, Beijing University of Chinese MedicineBeijing 100029, China
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical SciencesBeijing 100053, China
| | - Min Li
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical SciencesBeijing 100053, China
| | - Jingbo Zhao
- Department of Clinical Medicine, Aarhus UniversityAarhus 8200 N, Denmark
| | - Junling Li
- School of Traditional Chinese Medicine, Capital Medical UniversityBeijing 100069, China
| | - Guifang Liu
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical SciencesBeijing 100053, China
| | - Zhong Zhen
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical SciencesBeijing 100053, China
| | - Yang Cao
- Graduate School, Beijing University of Chinese MedicineBeijing 100029, China
| | - Hans Gregersen
- Bioengineering College of Chongqing UniversityChongqing 400044, China
- GIOME, Department of Surgery, Chinese University of Hong Kong and Prince of Wales HospitalShatin, Hong Kong
| | - Xiaolin Tong
- Department of Endocrinology, Guang’anmen Hospital, China Academy of Chinese Medical SciencesBeijing 100053, China
| |
Collapse
|
8
|
Amin H, Maccione A, Marinaro F, Zordan S, Nieus T, Berdondini L. Electrical Responses and Spontaneous Activity of Human iPS-Derived Neuronal Networks Characterized for 3-month Culture with 4096-Electrode Arrays. Front Neurosci 2016; 10:121. [PMID: 27065786 PMCID: PMC4811967 DOI: 10.3389/fnins.2016.00121] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 03/11/2016] [Indexed: 12/20/2022] Open
Abstract
The recent availability of human induced pluripotent stem cells (hiPSCs) holds great promise as a novel source of human-derived neurons for cell and tissue therapies as well as for in vitro drug screenings that might replace the use of animal models. However, there is still a considerable lack of knowledge on the functional properties of hiPSC-derived neuronal networks, thus limiting their application. Here, upon optimization of cell culture protocols, we demonstrate that both spontaneous and evoked electrical spiking activities of these networks can be characterized on-chip by taking advantage of the resolution provided by CMOS multielectrode arrays (CMOS-MEAs). These devices feature a large and closely-spaced array of 4096 simultaneously recording electrodes and multi-site on-chip electrical stimulation. Our results show that networks of human-derived neurons can respond to electrical stimulation with a physiological repertoire of spike waveforms after 3 months of cell culture, a period of time during which the network undergoes the expression of developing patterns of spontaneous spiking activity. To achieve this, we have investigated the impact on the network formation and on the emerging network-wide functional properties induced by different biochemical substrates, i.e., poly-dl-ornithine (PDLO), poly-l-ornithine (PLO), and polyethylenimine (PEI), that were used as adhesion promoters for the cell culture. Interestingly, we found that neuronal networks grown on PDLO coated substrates show significantly higher spontaneous firing activity, reliable responses to low-frequency electrical stimuli, and an appropriate level of PSD-95 that may denote a physiological neuronal maturation profile and synapse stabilization. However, our results also suggest that even 3-month culture might not be sufficient for human-derived neuronal network maturation. Taken together, our results highlight the tight relationship existing between substrate coatings and emerging network properties, i.e., spontaneous activity, responsiveness, synapse formation and maturation. Additionally, our results provide a baseline on the functional properties expressed over 3 months of network development for a commercially available line of hiPSC-derived neurons. This is a first step toward the development of functional pre-clinical assays to test pharmaceutical compounds on human-derived neuronal networks with CMOS-MEAs.
Collapse
Affiliation(s)
- Hayder Amin
- NetS3 Laboratory, Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia (IIT) Genova, Italy
| | - Alessandro Maccione
- NetS3 Laboratory, Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia (IIT) Genova, Italy
| | - Federica Marinaro
- Neurobiology of miRNA Laboratory, Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia (IIT) Genova, Italy
| | - Stefano Zordan
- NetS3 Laboratory, Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia (IIT) Genova, Italy
| | - Thierry Nieus
- NetS3 Laboratory, Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia (IIT) Genova, Italy
| | - Luca Berdondini
- NetS3 Laboratory, Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia (IIT) Genova, Italy
| |
Collapse
|
9
|
Fischer MJM, McNaughton PA. How anchoring proteins shape pain. Pharmacol Ther 2014; 143:316-22. [PMID: 24727631 DOI: 10.1016/j.pharmthera.2014.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 03/28/2014] [Indexed: 11/29/2022]
Abstract
Cellular responsiveness to external stimuli can be altered by extracellular mediators which activate membrane receptors, in turn signalling to the intracellular space via calcium, cyclic nucleotides, membrane lipids or enzyme activity. These signalling events trigger a cascade leading to an effector which can be a channel, an enzyme or a transcription factor. The effectiveness of these intracellular events is enhanced when they are maintained in close proximity by anchoring proteins, which assemble complexes of signalling molecules such as kinases together with their targets, and in this way enhance both the speed and the precision of intracellular signalling. The A kinase anchoring protein (AKAP) family are adaptor proteins originally named for their ability to associate Protein Kinase A and its targets, but several other enzymes bound by AKAPs have now been found and a wide variety of target structures has been described. This review provides an overview of anchoring proteins involved in pain signalling. The key anchoring proteins and their ion channel targets in primary sensory neurons responding to painful stimuli (nociceptors) are discussed.
Collapse
Affiliation(s)
- Michael J M Fischer
- Institute of Physiology and Pathophysiology, FAU Erlangen-Nürnberg, Germany.
| | - Peter A McNaughton
- Wolfson Centre for Age-Related Research, Hodgkin Building, King's College London, London SE1 1UH, UK
| |
Collapse
|
10
|
Fang ZH, Lee CH, Seo MK, Cho H, Lee JG, Lee BJ, Park SW, Kim YH. Effect of treadmill exercise on the BDNF-mediated pathway in the hippocampus of stressed rats. Neurosci Res 2013; 76:187-94. [DOI: 10.1016/j.neures.2013.04.005] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 04/26/2013] [Accepted: 04/26/2013] [Indexed: 01/08/2023]
|
11
|
Protein tyrosine phosphatases PTPδ, PTPσ, and LAR: presynaptic hubs for synapse organization. Trends Neurosci 2013; 36:522-34. [PMID: 23835198 DOI: 10.1016/j.tins.2013.06.002] [Citation(s) in RCA: 204] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 06/03/2013] [Accepted: 06/11/2013] [Indexed: 02/04/2023]
Abstract
Synapse development requires differentiation of presynaptic neurotransmitter release sites and postsynaptic receptive apparatus coordinated by synapse organizing proteins. In addition to the well-characterized neurexins, recent studies identified presynaptic type IIa receptor-type protein tyrosine phosphatases (RPTPs) as mediators of presynaptic differentiation and triggers of postsynaptic differentiation, thus extending the roles of RPTPs from axon outgrowth and guidance. Similarly to neurexins, RPTPs exist in multiple isoforms generated by alternative splicing that interact in a splice-selective code with diverse postsynaptic partners. The parallel RPTP and neurexin hub design facilitates synapse self-assembly through cooperation, pairs presynaptic similarity with postsynaptic diversity, and balances excitation with inhibition. Upon mutation of individual genes in neuropsychiatric disorders, imbalance of this synaptic organizing network may contribute to impaired cognitive function.
Collapse
|
12
|
Balasubramanian SK, Poh KW, Ong CN, Kreyling WG, Ong WY, Yu LE. The effect of primary particle size on biodistribution of inhaled gold nano-agglomerates. Biomaterials 2013; 34:5439-52. [PMID: 23639527 DOI: 10.1016/j.biomaterials.2013.03.080] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 03/27/2013] [Indexed: 01/15/2023]
Abstract
Airborne engineered nanoparticles undergo agglomeration, and careful distinction must be made between primary and agglomerate size of particles, when assessing their health effects. This study compares the effects on rats undergoing 15-day inhalation exposure to airborne agglomerates of gold nanoparticles (AuNPs) of similar size distribution and number concentration (1 × 10(6) particles/cm(3)), but two different primary diameters of 7 nm or 20 nm. Inhalation of agglomerates containing 7-nm AuNPs resulted in highest deposition by mass concentration in the lungs, followed by brain regions including the olfactory bulb, hippocampus, striatum, frontal cortex, entorhinal cortex, septum, cerebellum; aorta, esophagus, and kidney. Eight organs/tissues especially the brain retained greater mass concentration of Au after inhalation exposure to agglomerates of 7-nm than 20-nm AuNPs. Macrophage mediated escalation followed by fecal excretion is the major pathway of clearing inhaled AuNPs in the lungs. Microarray analyses of the hippocampus showed mostly downregulated genes, related to the cytoskeleton and neurite outgrowth. Together, results in this study indicate disintegration of nanosized agglomerates after inhalation and show impact of primary size of particles on subsequent biodistribution.
Collapse
Affiliation(s)
- Suresh K Balasubramanian
- Department of Civil and Environmental Engineering, National University of Singapore, Singapore 119260, Singapore
| | | | | | | | | | | |
Collapse
|
13
|
Ivanov A, Esclapez M, Ferhat L. Role of drebrin A in dendritic spine plasticity and synaptic function: Implications in neurological disorders. Commun Integr Biol 2011; 2:268-70. [PMID: 19641748 DOI: 10.4161/cib.2.3.8166] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2009] [Accepted: 02/13/2009] [Indexed: 11/19/2022] Open
Abstract
Drebrin A is one of the most abundant neuron-specific binding proteins of F-actin and its expression is increased in parallel with synapse formation. Drebrin A is particularly concentrated in dendritic spines, postsynaptic sides of excitatory glutamatergic synapses. More recently, Ferhat and colleagues reported the functional role of drebrin A in regulating synaptic transmission. Indeed, our study showed that overexpression of drebrin A induced an increase of glutamatergic but not GABAergic synapses and resulted in the alteration of the normal excitatory-inhibitory ratio in favor of excitation in mature hippocampal neurons. Downregulation of drebrin A expression by antisense oligonucleotides resulted in the decrease of both miniature- glutamatergic and GABAergic synaptic activities without affecting the excitatory-inhibitory ratio. Studies performed in heterologous cells revealed that drebrin A reorganized the actin filaments and stabilized them and that these effects are depend upon its actin-binding domain. These results suggest that drebrin A regulates dendritic spine morphology, size and density, presumably via regulation of actin cytoskeleton remodeling and dynamics. These data demonstrate for the first time that an actin-binding protein such as drebrin A regulates both glutamatergic and GABAergic synaptic transmissions, probably through an increase of active synaptic site density for glutamatergic transmission and through homeostatic mechanisms for the GABAergic one.It is appealing to suggest that abnormalities in the expression of drebrin A may result in aberrant synapse development and/or loss of synapses leading to synaptic dysfunction, which underlies cognitive impairment accompanying neurological disorders such as Alzheimer's disease, Down syndrome as well as normal aging.
Collapse
Affiliation(s)
- Anton Ivanov
- INSERM U 751; Université d'Aix-Marseille; Hôpital de la Timone; Marseille, France
| | | | | |
Collapse
|
14
|
Neuregulin 1 promotes excitatory synapse development and function in GABAergic interneurons. J Neurosci 2011; 31:15-25. [PMID: 21209185 DOI: 10.1523/jneurosci.2538-10.2011] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Neuregulin 1 (NRG1) and its receptor ErbB4 are both susceptibility genes of schizophrenia. However, little is known about the underlying mechanisms of their malfunction. Although ErbB4 is enriched in GABAergic interneurons, the role of NRG1 in excitatory synapse formation in these neurons remains poorly understood. We showed that NRG1 increased both the number and size of PSD-95 puncta and the frequency and amplitude of miniature EPSCs (mEPSCs) in GABAergic interneurons, indicating that NRG1 stimulates the formation of new synapses and strengthens existing synapses. In contrast, NRG1 treatment had no effect on either the number or size of excitatory synapses in glutamatergic neurons, suggesting its synaptogenic effect is specific to GABAergic interneurons. Ecto-ErbB4 treatment diminished both the number and size of excitatory synapses, suggesting that endogenous NRG1 may be critical for basal synapse formation. NRG1 could stimulate the stability of PSD-95 in the manner that requires tyrosine kinase activity of ErbB4. Finally, deletion of ErbB4 in parvalbumin-positive interneurons led to reduced frequency and amplitude of mEPSCs, providing in vivo evidence that ErbB4 is important in excitatory synaptogenesis in interneurons. Together, our findings suggested a novel synaptogenic role of NRG1 in excitatory synapse development, possibly via stabilizing PSD-95, and this effect is specific to GABAergic interneurons. In light of the association of the genes of both NRG1 and ErbB4 with schizophrenia and dysfunction of GABAergic system in this disorder, these results provide insight into its potential pathological mechanism.
Collapse
|
15
|
Moscato EH, Jain A, Peng X, Hughes EG, Dalmau J, Balice-Gordon RJ. Mechanisms underlying autoimmune synaptic encephalitis leading to disorders of memory, behavior and cognition: insights from molecular, cellular and synaptic studies. Eur J Neurosci 2010; 32:298-309. [PMID: 20646055 DOI: 10.1111/j.1460-9568.2010.07349.x] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Recently, several novel, potentially lethal and treatment-responsive syndromes that affect hippocampal and cortical function have been shown to be associated with auto-antibodies against synaptic antigens, notably glutamate or GABA-B receptors. Patients with these auto-antibodies, sometimes associated with teratomas and other neoplasms, present with psychiatric symptoms, seizures, memory deficits and decreased levels of consciousness. These symptoms often improve dramatically after immunotherapy or tumor resection. Here we review studies of the cellular and synaptic effects of these antibodies in hippocampal neurons in vitro and preliminary work in rodent models. Our work suggests that patient antibodies lead to rapid and reversible removal of neurotransmitter receptors from synaptic sites, leading to changes in synaptic and circuit function that in turn are likely to lead to behavioral deficits. We also discuss several of the many questions raised by these and related disorders. Determining the mechanisms underlying these novel anti-neurotransmitter receptor encephalopathies will provide insights into the cellular and synaptic bases of the memory and cognitive deficits that are hallmarks of these disorders, and potentially suggest avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Emilia H Moscato
- Department of Neuroscience, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6074, USA
| | | | | | | | | | | |
Collapse
|
16
|
Gatto CL, Broadie K. Genetic controls balancing excitatory and inhibitory synaptogenesis in neurodevelopmental disorder models. Front Synaptic Neurosci 2010; 2:4. [PMID: 21423490 PMCID: PMC3059704 DOI: 10.3389/fnsyn.2010.00004] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Accepted: 05/14/2010] [Indexed: 11/24/2022] Open
Abstract
Proper brain function requires stringent balance of excitatory and inhibitory synapse formation during neural circuit assembly. Mutation of genes that normally sculpt and maintain this balance results in severe dysfunction, causing neurodevelopmental disorders including autism, epilepsy and Rett syndrome. Such mutations may result in defective architectural structuring of synaptic connections, molecular assembly of synapses and/or functional synaptogenesis. The affected genes often encode synaptic components directly, but also include regulators that secondarily mediate the synthesis or assembly of synaptic proteins. The prime example is Fragile X syndrome (FXS), the leading heritable cause of both intellectual disability and autism spectrum disorders. FXS results from loss of mRNA-binding FMRP, which regulates synaptic transcript trafficking, stability and translation in activity-dependent synaptogenesis and plasticity mechanisms. Genetic models of FXS exhibit striking excitatory and inhibitory synapse imbalance, associated with impaired cognitive and social interaction behaviors. Downstream of translation control, a number of specific synaptic proteins regulate excitatory versus inhibitory synaptogenesis, independently or combinatorially, and loss of these proteins is also linked to disrupted neurodevelopment. The current effort is to define the cascade of events linking transcription, translation and the role of specific synaptic proteins in the maintenance of excitatory versus inhibitory synapses during neural circuit formation. This focus includes mechanisms that fine-tune excitation and inhibition during the refinement of functional synaptic circuits, and later modulate this balance throughout life. The use of powerful new genetic models has begun to shed light on the mechanistic bases of excitation/inhibition imbalance for a range of neurodevelopmental disease states.
Collapse
Affiliation(s)
- Cheryl L. Gatto
- Departments of Biological Sciences, Cell and Developmental Biology, Kennedy Center for Research on Human Development, Vanderbilt UniversityNashville, TN, USA
| | - Kendal Broadie
- Departments of Biological Sciences, Cell and Developmental Biology, Kennedy Center for Research on Human Development, Vanderbilt UniversityNashville, TN, USA
| |
Collapse
|
17
|
Neuroligin-2 deletion selectively decreases inhibitory synaptic transmission originating from fast-spiking but not from somatostatin-positive interneurons. J Neurosci 2009; 29:13883-97. [PMID: 19889999 DOI: 10.1523/jneurosci.2457-09.2009] [Citation(s) in RCA: 131] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Neuroligins are cell adhesion molecules involved in synapse formation and/or function. Neurons express four neuroligins (NL1-NL4), of which NL1 is specific to excitatory and NL2 to inhibitory synapses. Excitatory and inhibitory synapses include numerous subtypes. However, it is unknown whether NL1 performs similar functions in all excitatory and NL2 in all inhibitory synapses, or whether they regulate the formation and/or function of specific subsets of synapses. To address this central question, we performed paired recordings in primary somatosensory cortex of mice lacking NL1 or NL2. Using this system, we examined neocortical microcircuits formed by reciprocal synapses between excitatory neurons and two subtypes of inhibitory interneurons, namely, fast-spiking and somatostatin-positive interneurons. We find that the NL1 deletion had little effect on inhibitory synapses, whereas the NL2 deletion decreased (40-50%) the unitary (cell-to-cell) IPSC amplitude evoked from single fast-spiking interneurons. Strikingly, the NL2 deletion had no effect on IPSC amplitude evoked from single somatostatin-positive inhibitory interneurons. Moreover, the frequency of unitary synaptic connections between individual fast-spiking and somatostatin-positive interneurons and excitatory neurons was unchanged. The decrease in unitary IPSC amplitude originating from fast-spiking interneurons in NL2-deficient mice was due to a multiplicative and uniform downscaling of the amplitude distribution, which in turn was mediated by a decrease in both synaptic quantal amplitude and quantal content, the latter inferred from an increase in the coefficient of variation. Thus, NL2 is not necessary for establishing unitary inhibitory synaptic connections but is selectively required for "scaling up" unitary connections originating from a subset of interneurons.
Collapse
|
18
|
Ivanov A, Esclapez M, Pellegrino C, Shirao T, Ferhat L. Drebrin A regulates dendritic spine plasticity and synaptic function in mature cultured hippocampal neurons. J Cell Sci 2009; 122:524-34. [PMID: 19174472 DOI: 10.1242/jcs.033464] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Drebrin A, one of the most abundant neuron-specific F-actin-binding proteins, is found exclusively in dendrites and is particularly concentrated in dendritic spines receiving excitatory inputs. We investigated the role of drebrin A in synaptic transmission and found that overexpression of drebrin A augmented the glutamatergic synaptic transmission, probably through an increase of active synaptic site density. Interestingly, overexpression of drebrin A also affected the frequency, amplitude and kinetics of miniature inhibitory postsynaptic currents (mIPSCs), despite the fact that GABAergic synapse density and transmission efficacy were not modified. Downregulation of drebrin A led to a decrease of both glutamatergic and GABAergic synaptic activity. In heterologous cells, drebrin A reorganized and stabilized F-actin and these effects were mediated by its actin-binding domain. Thus, drebrin A might regulate dendritic spine morphology via regulation of actin cytoskeleton remodeling and dynamics. Our data demonstrate for the first time that drebrin A modulates glutamatergic and GABAergic synaptic activities.
Collapse
Affiliation(s)
- Anton Ivanov
- INMED/INSERM U29, Parc Scientifique de Luminy, 13273, Marseille, France
| | | | | | | | | |
Collapse
|
19
|
Keith D, El-Husseini A. Excitation Control: Balancing PSD-95 Function at the Synapse. Front Mol Neurosci 2008; 1:4. [PMID: 18946537 PMCID: PMC2526002 DOI: 10.3389/neuro.02.004.2008] [Citation(s) in RCA: 147] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2008] [Accepted: 01/30/2008] [Indexed: 01/12/2023] Open
Abstract
Excitability of individual neurons dictates the overall excitation in specific brain circuits. This process is thought to be regulated by molecules that regulate synapse number, morphology and strength. Neuronal excitation is also influenced by the amounts of neurotransmitter receptors and signaling molecules retained at particular synaptic sites. Recent studies revealed a key role for PSD-95, a scaffolding molecule enriched at glutamatergic synapses, in modulation of clustering of several neurotransmitter receptors, adhesion molecules, ion channels, cytoskeletal elements and signaling molecules at postsynaptic sites. In this review we will highlight mechanisms that control targeting of PSD-95 at the synapse, and discuss how this molecule influences the retention and clustering of diverse synaptic proteins to regulate synaptic structure and strength. We will also discuss how PSD-95 may maintain a balance between excitation and inhibition in the brain and how alterations in this balance may contribute to neuropsychiatric disorders.
Collapse
Affiliation(s)
- Dove Keith
- Department of Psychiatry and the Brain Research Centre, University of British Columbia Vancouver, BC, Canada
| | | |
Collapse
|
20
|
Keith D, El-Husseini A. Excitation Control: Balancing PSD-95 Function at the Synapse. Front Mol Neurosci 2008; 1:4. [PMID: 18946537 DOI: 10.3389/neuro.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2008] [Accepted: 01/30/2008] [Indexed: 05/19/2023] Open
Abstract
Excitability of individual neurons dictates the overall excitation in specific brain circuits. This process is thought to be regulated by molecules that regulate synapse number, morphology and strength. Neuronal excitation is also influenced by the amounts of neurotransmitter receptors and signaling molecules retained at particular synaptic sites. Recent studies revealed a key role for PSD-95, a scaffolding molecule enriched at glutamatergic synapses, in modulation of clustering of several neurotransmitter receptors, adhesion molecules, ion channels, cytoskeletal elements and signaling molecules at postsynaptic sites. In this review we will highlight mechanisms that control targeting of PSD-95 at the synapse, and discuss how this molecule influences the retention and clustering of diverse synaptic proteins to regulate synaptic structure and strength. We will also discuss how PSD-95 may maintain a balance between excitation and inhibition in the brain and how alterations in this balance may contribute to neuropsychiatric disorders.
Collapse
Affiliation(s)
- Dove Keith
- Department of Psychiatry and the Brain Research Centre, University of British Columbia Vancouver, BC, Canada
| | | |
Collapse
|
21
|
Chubykin AA, Atasoy D, Etherton MR, Brose N, Kavalali ET, Gibson JR, Südhof TC. Activity-dependent validation of excitatory versus inhibitory synapses by neuroligin-1 versus neuroligin-2. Neuron 2007; 54:919-31. [PMID: 17582332 PMCID: PMC3738748 DOI: 10.1016/j.neuron.2007.05.029] [Citation(s) in RCA: 450] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2006] [Revised: 09/29/2006] [Accepted: 05/31/2007] [Indexed: 01/08/2023]
Abstract
Neuroligins enhance synapse formation in vitro, but surprisingly are not required for the generation of synapses in vivo. We now show that in cultured neurons, neuroligin-1 overexpression increases excitatory, but not inhibitory, synaptic responses, and potentiates synaptic NMDAR/AMPAR ratios. In contrast, neuroligin-2 overexpression increases inhibitory, but not excitatory, synaptic responses. Accordingly, deletion of neuroligin-1 in knockout mice selectively decreases the NMDAR/AMPAR ratio, whereas deletion of neuroligin-2 selectively decreases inhibitory synaptic responses. Strikingly, chronic inhibition of NMDARs or CaM-Kinase II, which signals downstream of NMDARs, suppresses the synapse-boosting activity of neuroligin-1, whereas chronic inhibition of general synaptic activity suppresses the synapse-boosting activity of neuroligin-2. Taken together, these data indicate that neuroligins do not establish, but specify and validate, synapses via an activity-dependent mechanism, with different neuroligins acting on distinct types of synapses. This hypothesis reconciles the overexpression and knockout phenotypes and suggests that neuroligins contribute to the use-dependent formation of neural circuits.
Collapse
Affiliation(s)
- Alexander A Chubykin
- Department of Neuroscience, The University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Gingras J, Rassadi S, Cooper E, Ferns M. Synaptic transmission is impaired at neuronal autonomic synapses in agrin-null mice. Dev Neurobiol 2007; 67:521-34. [PMID: 17443806 DOI: 10.1002/dneu.20304] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neuronal synapse formation is a multistep process regulated by several pre- and postsynaptic adhesion and signaling proteins. Recently, we found that agrin acts as one such synaptogenic factor at neuronal synapses in the PNS by demonstrating that structural synapse formation is impaired in the superior cervical ganglia (SCG) of z+ agrin-deficient mice and in SCG cultures derived from those animals. Here, we tested whether synaptic function is defective in agrin-null (AGD-/-) ganglia and began to define agrin's mechanism of action. Our electrophysiological recordings of compound action potentials showed that presynaptic stimulation evoked action potentials in approximately 40% of AGD-/- ganglionic neurons compared to 90% of wild-type neurons; moreover, transmission could not be potentiated as in wild-type or z+ agrin-deficient ganglia. Intracellular recordings also showed that nerve-evoked excitatory postsynaptic potentials in AGD-/- neurons were only 1/3 the size of those in wild-type neurons and mostly subthreshold. Consistent with these defects in transmission, we found an approximately 40-50% decrease in synapse number in AGD-/- ganglia and cultures, and decreased levels of differentiation at the residual synapses in culture. Furthermore, surface levels of acetylcholine receptors (AChRs) were equivalent in cultured AGD-/- and wild-type neurons, and depolarization reduced the synaptic localization of AChRs in AGD-/- but not wild-type neurons. These findings provide the first direct demonstration that agrin is required for proper structural and functional development of an interneuronal synapse in vivo. Moreover, they suggest a novel role for agrin, in stabilizing the postsynaptic density of nAChR at nascent neuronal synapses.
Collapse
Affiliation(s)
- Jacinthe Gingras
- Center for Research in Neuroscience, McGill University Health Center, Montreal, QC, H3G 1A4, Canada
| | | | | | | |
Collapse
|
23
|
Dong N, Qi J, Chen G. Molecular reconstitution of functional GABAergic synapses with expression of neuroligin-2 and GABAA receptors. Mol Cell Neurosci 2007; 35:14-23. [PMID: 17336090 DOI: 10.1016/j.mcn.2007.01.013] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2006] [Revised: 12/31/2006] [Accepted: 01/24/2007] [Indexed: 11/18/2022] Open
Abstract
Trans-synaptic cell adhesion molecules neuroligins and neurexins play an important role in promoting central synapse formation. We report here a molecular reconstruction of functional GABAergic synapses in non-neuronal cells with the coexpression of postsynaptic cell adhesion molecule neuroligin-2 (NL-2) and GABA(A) receptors. HEK 293T cells were co-transfected with GABA(A) receptors and NL-2 or its homologue neuroligin-1 (NL-1), and then cocultured with hypothalamic cultures which are enriched with GABAergic neurons. Both spontaneous and action potential-evoked GABAergic events were readily detected in HEK 293T cells coexpressing GABA(A) receptors with NL-2, but not with NL-1. Aggregating NL-2 with specific antibodies in live cells resulted in coaggregation of GABA(A) receptors. Expression of NL-2 in HEK 293T cells also induced stronger GABAergic presynaptic differentiation than that of NL-1 in neuronal cocultures. These results suggest that NL-2 may potentially serve as a central organizer for GABAergic synapse assembly by interacting with both presynaptic neurexins and postsynaptic GABA(A) receptors.
Collapse
Affiliation(s)
- Ning Dong
- Department of Biology, The Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | | | | |
Collapse
|
24
|
Arancibia-Carcamo IL, Moss SJ. Molecular organization and assembly of the central inhibitory postsynapse. Results Probl Cell Differ 2006; 43:25-47. [PMID: 17068966 DOI: 10.1007/400_017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
gamma-Amino butyric acid type A (GABAA) receptors are the major sites of fast synaptic inhibition in the brain. GABAA receptors play an important role in regulating neuronal excitability and in addition have been implicated in numerous neurological disorders. In order to understand synaptic inhibition it is important to comprehend the cellular mechanisms, that neurons utilize to regulate the accumulation and regulation of GABAA receptors at postsynaptic inhibitory specializations. Over the past decade a number of GABAA receptor interacting proteins have been identified allowing us to further understand the trafficking, targeting and clustering of these receptors as well as the regulation of receptor stability. In the following review we examine the proteins identified as GABAA receptor binding partners and other components of the inhibitory postsynaptic scaffold, and how they contribute to the construction of inhibitory synapses and the dynamic modulation of synaptic inhibition.
Collapse
|
25
|
Badcock C, Crespi B. Imbalanced genomic imprinting in brain development: an evolutionary basis for the aetiology of autism. J Evol Biol 2006; 19:1007-32. [PMID: 16780503 DOI: 10.1111/j.1420-9101.2006.01091.x] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
We describe a new hypothesis for the development of autism, that it is driven by imbalances in brain development involving enhanced effects of paternally expressed imprinted genes, deficits of effects from maternally expressed genes, or both. This hypothesis is supported by: (1) the strong genomic-imprinting component to the genetic and developmental mechanisms of autism, Angelman syndrome, Rett syndrome and Turner syndrome; (2) the core behavioural features of autism, such as self-focused behaviour, altered social interactions and language, and enhanced spatial and mechanistic cognition and abilities, and (3) the degree to which relevant brain functions and structures are altered in autism and related disorders. The imprinted brain theory of autism has important implications for understanding the genetic, epigenetic, neurological and cognitive bases of autism, as ultimately due to imbalances in the outcomes of intragenomic conflict between effects of maternally vs. paternally expressed genes.
Collapse
Affiliation(s)
- C Badcock
- Department of Sociology, London School of Economics, London, UK
| | | |
Collapse
|
26
|
Butkevich IP, Barr GA, Mikhailenko VA, Otellin VA. Increased formalin-induced pain and expression of fos neurons in the lumbar spinal cord of prenatally stressed infant rats. Neurosci Lett 2006; 403:222-6. [PMID: 16782271 DOI: 10.1016/j.neulet.2006.04.059] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2006] [Revised: 04/26/2006] [Accepted: 04/27/2006] [Indexed: 11/26/2022]
Abstract
When pregnant dams are stressed, there is a resultant alteration in brain development and behavior in their offspring. Prior work has shown increased nociceptive responses in adolescent or adult rats born of stressed dams. However, the age at which those changes first occur is not known. The aim of the present study was to evaluate the effect of prenatal stress on pain sensitivity in the formalin test in 7-day-old rats, behaviorally and by fos-like immunoreactivity (Fos-LI) in the lumbar spinal cord dorsal horn. The behavioral response to intraplantar injection of formalin is represented by two nociceptive phases separated by an interphase during which nociceptive responses decrease; the interphase is not seen until the start of the third postnatal week and appears as descending inhibitory monoaminergic systems develop. Prenatally stressed infants showed increased nociceptive responses in the second, tonic phase and a large increase in the number of formalin-induced Fos-LI neurons in the lumbar dorsal horn, a result consistent with the behavioral data. The increased nociception in prenatally stressed 7-day-old pups may be associated with the decrease in the intensity of serotonin-like immunoreactivity and density of serotonergic cells in the lumbar spinal cord dorsal horn and the dorsal raphe nucleus reported earlier.
Collapse
Affiliation(s)
- Irina P Butkevich
- Laboratory of Ontogeny of Nervous System, I.P. Pavlov Institute of Physiology, The Russian Academy of Sciences, St. Petersburg 199034, Russia.
| | | | | | | |
Collapse
|
27
|
Calabrese B, Wilson MS, Halpain S. Development and regulation of dendritic spine synapses. Physiology (Bethesda) 2006; 21:38-47. [PMID: 16443821 DOI: 10.1152/physiol.00042.2005] [Citation(s) in RCA: 175] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Dendritic spines are small protrusions from neuronal dendrites that form the postsynaptic component of most excitatory synapses in the brain. They play critical roles in synaptic transmission and plasticity. Recent advances in imaging and molecular technologies reveal that spines are complex, dynamic structures that contain a dense array of cytoskeletal, transmembrane, and scaffolding molecules. Several neurological and psychiatric disorders exhibit dendritic spine abnormalities.
Collapse
Affiliation(s)
- Barbara Calabrese
- Department of Cell Biology and Institute for Childhood and Neglected Diseases, The Scripps Research Institute, La Jolla, California, USA
| | | | | |
Collapse
|
28
|
Dean C, Dresbach T. Neuroligins and neurexins: linking cell adhesion, synapse formation and cognitive function. Trends Neurosci 2005; 29:21-9. [PMID: 16337696 DOI: 10.1016/j.tins.2005.11.003] [Citation(s) in RCA: 246] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2005] [Revised: 09/02/2005] [Accepted: 11/10/2005] [Indexed: 12/20/2022]
Abstract
Cell adhesion represents the most direct way of coordinating synaptic connectivity in the brain. Recent evidence highlights the importance of a trans-synaptic interaction between postsynaptic neuroligins and presynaptic neurexins. These transmembrane molecules bind each other extracellularly to promote adhesion between dendrites and axons. This signals the recruitment of presynaptic and postsynaptic molecules to form a functional synapse. Remarkably, neuroligins alone can induce the formation of fully functional presynaptic terminals in contacting axons. Conversely, neurexins alone can induce postsynaptic differentiation and clustering of receptors in dendrites. Therefore, the neuroligin-neurexin interaction has the unique ability to act as a bi-directional trigger of synapse formation. Here, we review several recent studies that offer clues as to how these proteins form synapses and how they might function in the brain to establish and modify neuronal network properties and cognition.
Collapse
Affiliation(s)
- Camin Dean
- Department of Physiology, University of Wisconsin Medical School, Madison, WI 53706, USA.
| | | |
Collapse
|
29
|
Affiliation(s)
- Min Zhuo
- Department of Physiology, Faculty of Medicine, University of Toronto, Centre for the Study of Pain, Toronto, M5S 1A8, Canada.
| |
Collapse
|