51
|
Aberrant ventral dentate gyrus structure and function in trauma susceptible mice. Transl Psychiatry 2022; 12:502. [PMID: 36473832 PMCID: PMC9723770 DOI: 10.1038/s41398-022-02264-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 11/18/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) is a psychiatric disorder vulnerable individuals can develop following a traumatic event, whereas others are resilient. Enhanced insight into the mechanistic underpinnings contributing to these inter-individual differences in trauma susceptibility is key to improved treatment and prevention. Aberrant function of the hippocampal dentate gyrus (DG) may contribute to its psychopathology, with the dorsal DG potentially encoding trauma memory generalization and the ventral DG anxiety. Using a mouse model, we hypothesized that susceptibility to develop PTSD-like symptoms following trauma will be underpinned by aberrant DG structure and function. Mice were exposed to a traumatic event (unpredictable, inescapable foot shocks) and tested for PTSD-like symptomatology following recovery. In four independent experiments, DG neuronal morphology, synaptic protein gene and protein expression, and neuronal activity during trauma encoding and recall were assessed. Behaviorally, trauma-susceptible animals displayed increased anxiety-like behavior already prior to trauma, increased novelty-induced freezing, but no clear differences in remote trauma memory recall. Comparison of the ventral DG of trauma susceptible vs resilient mice revealed lower spine density, reduced expression of the postsynaptic protein homer1b/c gene and protein, a larger population of neurons active during trauma encoding, and a greater presence of somatostatin neurons. In contrast, the dorsal DG of trauma-susceptible animals did not differ in terms of spine density or gene expression but displayed more active neurons during trauma encoding and a lower amount of somatostatin neurons. Collectively, we here report on specific structural and functional changes in the ventral DG in trauma susceptible male mice.
Collapse
|
52
|
Liao M, Hu Y, Zhang Y, Wang K, Fang Q, Qi Y, Shen Y, Cheng H, Fu X, Tang M, Sun S, Gao X, Chai R. 3D Ti 3C 2T x MXene-Matrigel with Electroacoustic Stimulation to Promote the Growth of Spiral Ganglion Neurons. ACS NANO 2022; 16:16744-16756. [PMID: 36222600 PMCID: PMC9620407 DOI: 10.1021/acsnano.2c06306] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Cochlear implantation has become the most effective treatment method for patients with profound and total hearing loss. However, its therapeutic efficacy is dependent on the number and normal physiological function of cochlear implant-targeted spiral ganglion neurons (SGNs). Electrical stimulation can be used as an effective cue to regulate the morphology and function of excitatory cells. Therefore, it is important to develop an efficient cochlear implant electroacoustic stimulation (EAS) system to study the behavior of SGNs. In this work, we present an electrical stimulation system constructed by combining a cochlear implant and a conductive Ti3C2Tx MXene-matrigel hydrogel. SGNs were cultured in the Ti3C2Tx MXene-matrigel hydrogel and exposed to electrical stimulation transduced by the cochlear implant. It was demonstrated that low-frequency stimulation promoted the growth cone development and neurite outgrowth of SGNs as well as signal transmission between cells. This work may have potential value for the clinical application of the Ti3C2Tx MXene hydrogel to optimize the postoperative listening effect of cochlear implantation and benefit people with sensorineural hearing loss.
Collapse
Affiliation(s)
- Menghui Liao
- State
Key Laboratory of Bioelectronics, Department of Otolaryngology Head
and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology,
Advanced Institute for Life and Health, Jiangsu Province High-Tech
Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, Jiangsu 210096, China
- Department
of Otorhinolaryngology−Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, China
| | - Yangnan Hu
- State
Key Laboratory of Bioelectronics, Department of Otolaryngology Head
and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology,
Advanced Institute for Life and Health, Jiangsu Province High-Tech
Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, Jiangsu 210096, China
- Department
of Otorhinolaryngology−Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, China
| | - Yuhua Zhang
- State
Key Laboratory of Bioelectronics, Department of Otolaryngology Head
and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology,
Advanced Institute for Life and Health, Jiangsu Province High-Tech
Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, Jiangsu 210096, China
| | - Kaichen Wang
- Chien-Shiung
Wu College, Southeast University, Nanjing, Jiangsu 210096, China
| | - Qiaojun Fang
- State
Key Laboratory of Bioelectronics, Department of Otolaryngology Head
and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology,
Advanced Institute for Life and Health, Jiangsu Province High-Tech
Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, Jiangsu 210096, China
| | - Yanru Qi
- State
Key Laboratory of Bioelectronics, Department of Otolaryngology Head
and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology,
Advanced Institute for Life and Health, Jiangsu Province High-Tech
Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, Jiangsu 210096, China
| | - Yingbo Shen
- Chien-Shiung
Wu College, Southeast University, Nanjing, Jiangsu 210096, China
| | - Hong Cheng
- State
Key Laboratory of Bioelectronics, Department of Otolaryngology Head
and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology,
Advanced Institute for Life and Health, Jiangsu Province High-Tech
Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, Jiangsu 210096, China
| | - Xiaolong Fu
- State
Key Laboratory of Bioelectronics, Department of Otolaryngology Head
and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology,
Advanced Institute for Life and Health, Jiangsu Province High-Tech
Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, Jiangsu 210096, China
| | - Mingliang Tang
- State
Key Laboratory of Bioelectronics, Department of Otolaryngology Head
and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology,
Advanced Institute for Life and Health, Jiangsu Province High-Tech
Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, Jiangsu 210096, China
- Institute for Cardiovascular Science and Department of Cardiovascular Surgery of the First Affiliated
Hospital, Medical College, Soochow University, Suzhou, Jiangsu 215000, China
- Co-Innovation
Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Shan Sun
- ENT
Institute and Department
of Otorhinolaryngology, Eye and ENT Hospital, State Key Laboratory
of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200031, China
| | - Xia Gao
- Department
of Otorhinolaryngology−Head and Neck Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, China
| | - Renjie Chai
- State
Key Laboratory of Bioelectronics, Department of Otolaryngology Head
and Neck Surgery, Zhongda Hospital, School of Life Sciences and Technology,
Advanced Institute for Life and Health, Jiangsu Province High-Tech
Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, Jiangsu 210096, China
- Co-Innovation
Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
- Department
of Otolaryngology−Head and Neck Surgery, Sichuan Provincial
People’s Hospital, University of
Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
- Institute
for Stem Cell and Regeneration, Chinese
Academy of Science, Beijing 100101, China
- Beijing
Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing 100069, China
| |
Collapse
|
53
|
Lehr AB, Luboeinski J, Tetzlaff C. Neuromodulator-dependent synaptic tagging and capture retroactively controls neural coding in spiking neural networks. Sci Rep 2022; 12:17772. [PMID: 36273097 PMCID: PMC9588040 DOI: 10.1038/s41598-022-22430-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 10/14/2022] [Indexed: 01/19/2023] Open
Abstract
Events that are important to an individual's life trigger neuromodulator release in brain areas responsible for cognitive and behavioral function. While it is well known that the presence of neuromodulators such as dopamine and norepinephrine is required for memory consolidation, the impact of neuromodulator concentration is, however, less understood. In a recurrent spiking neural network model featuring neuromodulator-dependent synaptic tagging and capture, we study how synaptic memory consolidation depends on the amount of neuromodulator present in the minutes to hours after learning. We find that the storage of rate-based and spike timing-based information is controlled by the level of neuromodulation. Specifically, we find better recall of temporal information for high levels of neuromodulation, while we find better recall of rate-coded spatial patterns for lower neuromodulation, mediated by the selection of different groups of synapses for consolidation. Hence, our results indicate that in minutes to hours after learning, the level of neuromodulation may alter the process of synaptic consolidation to ultimately control which type of information becomes consolidated in the recurrent neural network.
Collapse
Affiliation(s)
- Andrew B. Lehr
- grid.7450.60000 0001 2364 4210Department of Computational Neuroscience, University of Göttingen, Göttingen, Germany ,grid.7450.60000 0001 2364 4210Bernstein Center for Computational Neuroscience, University of Göttingen, Göttingen, Germany ,grid.7450.60000 0001 2364 4210Department of Computational Synaptic Physiology, University of Göttingen, Göttingen, Germany
| | - Jannik Luboeinski
- grid.7450.60000 0001 2364 4210Department of Computational Neuroscience, University of Göttingen, Göttingen, Germany ,grid.7450.60000 0001 2364 4210Bernstein Center for Computational Neuroscience, University of Göttingen, Göttingen, Germany ,grid.7450.60000 0001 2364 4210Department of Computational Synaptic Physiology, University of Göttingen, Göttingen, Germany
| | - Christian Tetzlaff
- grid.7450.60000 0001 2364 4210Department of Computational Neuroscience, University of Göttingen, Göttingen, Germany ,grid.7450.60000 0001 2364 4210Bernstein Center for Computational Neuroscience, University of Göttingen, Göttingen, Germany ,grid.7450.60000 0001 2364 4210Department of Computational Synaptic Physiology, University of Göttingen, Göttingen, Germany
| |
Collapse
|
54
|
Abstract
Activation of Ca2+/calmodulin-dependent kinase II (CaMKII) plays a critical role in long-term potentiation (LTP), a long accepted cellular model for learning and memory. However, how LTP and memories survive the turnover of synaptic proteins, particularly CaMKII, remains a mystery. Here, we take advantage of the finding that constitutive Ca2+-independent CaMKII activity, acquired prior to slice preparation, provides a lasting memory trace at synapses. In slice culture, this persistent CaMKII activity, in the absence of Ca2+ stimulation, remains stable over a 2-wk period, well beyond the turnover of CaMKII protein. We propose that the nascent CaMKII protein present at 2 wk acquired its activity from preexisting active CaMKII molecules, which transferred their activity to newly synthesized CaMKII molecules and thus maintain the memory in the face of protein turnover.
Collapse
|
55
|
Wang X, Jin Y, Hao K. Computational Modeling of Structural Synaptic Plasticity in Echo State Networks. IEEE TRANSACTIONS ON CYBERNETICS 2022; 52:11254-11266. [PMID: 33760748 DOI: 10.1109/tcyb.2021.3060466] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Most existing studies on computational modeling of neural plasticity have focused on synaptic plasticity. However, regulation of the internal weights in the reservoir based on synaptic plasticity often results in unstable learning dynamics. In this article, a structural synaptic plasticity learning rule is proposed to train the weights and add or remove neurons within the reservoir, which is shown to be able to alleviate the instability of the synaptic plasticity, and to contribute to increase the memory capacity of the network as well. Our experimental results also reveal that a few stronger connections may last for a longer period of time in a constantly changing network structure, and are relatively resistant to decay or disruptions in the learning process. These results are consistent with the evidence observed in biological systems. Finally, we show that an echo state network (ESN) using the proposed structural plasticity rule outperforms an ESN using synaptic plasticity and three state-of-the-art ESNs on four benchmark tasks.
Collapse
|
56
|
Lyu J, Yao Z, Liang B, Liu Y, Zhang Y. Small protein complex prediction algorithm based on protein-protein interaction network segmentation. BMC Bioinformatics 2022; 23:405. [PMID: 36180820 PMCID: PMC9524060 DOI: 10.1186/s12859-022-04960-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 09/19/2022] [Indexed: 11/23/2022] Open
Abstract
Background Identifying protein complexes from protein-protein interaction network is one of significant tasks in the postgenome era. Protein complexes, none of which exceeds 10 in size play an irreplaceable role in life activities and are also a hotspot of scientific research, such as PSD-95, CD44, PKM2 and BRD4. And in MIPS, CYC2008, SGD, Aloy and TAP06 datasets, the proportion of small protein complexes is over 75%. But up to now, protein complex identification methods do not perform well in the field of small protein complexes. Results In this paper, we propose a novel method, called BOPS. It is a three-step procedure. Firstly, it calculates the balanced weights to replace the original weights. Secondly, it divides the graphs larger than MAXP until the original PPIN is divided into small PPINs. Thirdly, it enumerates the connected subset of each small PPINs, identifies potential protein complexes based on cohesion and removes those that are similar. Conclusions In four yeast PPINs, experimental results have shown that BOPS has an improvement of about 5% compared with the SOTA model. In addition, we constructed a weighted Homo sapiens PPIN based on STRINGdb and BioGRID, and BOPS gets the best result in it. These results give new insights into the identification of small protein complexes, and the weighted Homo sapiens PPIN provides more data for related research.
Collapse
Affiliation(s)
- Jiaqing Lyu
- School of Computer Science and Technology, Dalian University of Technology, Dalian, China
| | - Zhen Yao
- School of Chemical Engineering, Dalian University of Technology, Dalian, China
| | - Bing Liang
- School of Innovation and Entrepreneurship, Dalian University of Technology, Dalian, China.
| | - Yiwei Liu
- School of Innovation and Entrepreneurship, Dalian University of Technology, Dalian, China
| | - Yijia Zhang
- School of Information Science and Technology, Dalian Maritime University, Dalian, China.
| |
Collapse
|
57
|
Jing G, Zuo J, Fang Q, Yuan M, Xia Y, Jin Q, Liu Y, Wang Y, Zhang Z, Liu W, Wu X, Song X. Erbin protects against sepsis-associated encephalopathy by attenuating microglia pyroptosis via IRE1α/Xbp1s-Ca 2+ axis. J Neuroinflammation 2022; 19:237. [PMID: 36171629 PMCID: PMC9520943 DOI: 10.1186/s12974-022-02598-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 09/16/2022] [Indexed: 06/27/2024] Open
Abstract
Background Microglia pyroptosis-mediated neuroinflammation is thought to be the crucial pathogenesis of sepsis-associated encephalopathy (SAE). Erbin has been reported to be associated with various inflammatory diseases. However, the role of Erbin in SAE and the relationship between Erbin and microglia pyroptosis are unknown. In this study, we investigated the promising role and underlying molecular mechanism of Erbin in the regulation of microglia pyroptosis. Methods WT and Erbin knockout mice underwent cecum ligation perforation (CLP) to induce SAE. Primary mouse microglia and BV2 cells were treated with LPS/nigericin in vitro. Behavioral tests were performed to evaluate cognitive function. Nissl staining and transmission electron microscopy were used to assess histological and structural lesions. ELISA and qPCR were carried out to detect neuroinflammation. Western blot and immunofluorescence were used to analyze protein expression. Flow cytometry and confocal microscopy were utilized to observe the Ca2+ changes in the cytoplasm and endoplasmic reticulum (ER). To further explore the underlying mechanism, STF083010 was administered to block the IRE1α/Xbp1s pathway. Results Erbin deletion resulted in more pronounced neuronal damage and cognitive impairment in mice that underwent CLP. Erbin knockout promoted microglial pyroptosis and inflammatory cytokines secretion in vivo and in vitro, which was mediated by activation of the IRE1α/Xbp1s. Treatment with the selective inhibitor STF083010 significantly inhibited IRE1α/Xbp1s pathway activity, decreased intracytoplasmic Ca2+, attenuated microglial pyroptosis, reduced pro-inflammatory cytokine secretion, lessened neuronal damage, and improved cognitive function. Conclusions In SAE, Erbin inhibits IRE1/Xbp1s pathway activity and reduces the ER Ca2+ influx to the cytoplasm, reducing microglial pyroptosis. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02598-5.
Collapse
Affiliation(s)
- Guoqing Jing
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jing Zuo
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qing Fang
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Min Yuan
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yun Xia
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qiyan Jin
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yuping Liu
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yanlin Wang
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zongze Zhang
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wanhong Liu
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China.
| | - Xiaojing Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Xuemin Song
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
58
|
Amano R, Nakao M, Matsumiya K, Miwakeichi F. A computational model to explore how temporal stimulation patterns affect synapse plasticity. PLoS One 2022; 17:e0275059. [PMID: 36149886 PMCID: PMC9506666 DOI: 10.1371/journal.pone.0275059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 09/09/2022] [Indexed: 11/18/2022] Open
Abstract
Plasticity-related proteins (PRPs), which are synthesized in a synapse activation-dependent manner, are shared by multiple synapses to a limited spatial extent for a specific period. In addition, stimulated synapses can utilize shared PRPs through synaptic tagging and capture (STC). In particular, the phenomenon by which short-lived early long-term potentiation is transformed into long-lived late long-term potentiation using shared PRPs is called “late-associativity,” which is the underlying principle of “cluster plasticity.” We hypothesized that the competitive capture of PRPs by multiple synapses modulates late-associativity and affects the fate of each synapse in terms of whether it is integrated into a synapse cluster. We tested our hypothesis by developing a computational model to simulate STC, late-associativity, and the competitive capture of PRPs. The experimental results obtained using the model revealed that the number of competing synapses, timing of stimulation to each synapse, and basal PRP level in the dendritic compartment altered the effective temporal window of STC and influenced the conditions under which late-associativity occurs. Furthermore, it is suggested that the competitive capture of PRPs results in the selection of synapses to be integrated into a synapse cluster via late-associativity.
Collapse
Affiliation(s)
- Ryota Amano
- Graduate School of Information Sciences, Tohoku University, Sendai, Japan
- * E-mail:
| | - Mitsuyuki Nakao
- Graduate School of Information Sciences, Tohoku University, Sendai, Japan
| | | | - Fumikazu Miwakeichi
- Graduate School of Information Sciences, Tohoku University, Sendai, Japan
- Department of Statistical Modeling, The Institute of Statistical Mathematics, Tachikawa-Shi, Japan
| |
Collapse
|
59
|
Gore A, Yurina A, Yukevich-Mussomeli A, Nahmani M. Synaptic spinules are reliable indicators of excitatory presynaptic bouton size and strength and are ubiquitous components of excitatory synapses in CA1 hippocampus. Front Synaptic Neurosci 2022; 14:968404. [PMID: 36032419 PMCID: PMC9403541 DOI: 10.3389/fnsyn.2022.968404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/25/2022] [Indexed: 11/23/2022] Open
Abstract
Synaptic spinules are thin, finger-like projections from one neuron that become embedded within the presynaptic or postsynaptic compartments of another neuron. While spinules are conserved features of synapses across the animal kingdom, their specific function(s) remain unknown. Recent focused ion beam scanning electron microscopy (FIB-SEM) image volume analyses have demonstrated that spinules are embedded within ∼25% of excitatory boutons in primary visual cortex, yet the diversity of spinule sizes, origins, and ultrastructural relationships to their boutons remained unclear. To begin to uncover the function of synaptic spinules, we sought to determine the abundance, origins, and 3D ultrastructure of spinules within excitatory presynaptic spinule-bearing boutons (SBBs) in mammalian CA1 hippocampus and compare them with presynaptic boutons bereft of spinules (non-SBBs). Accordingly, we performed a comprehensive 3D analysis of every excitatory presynaptic bouton, their embedded spinules, and postsynaptic densities, within a 5 nm isotropic FIB-SEM image volume from CA1 hippocampus of an adult male rat. Surprisingly, we found that ∼74% of excitatory presynaptic boutons in this volume contained at least one spinule, suggesting they are fundamental components of excitatory synapses in CA1. In addition, we found that SBBs are 2.5-times larger and have 60% larger postsynaptic densities (PSDs) than non-SBBs. Moreover, synaptic spinules within SBBs are clearly differentiated into two groups: small clathrin-coated spinules, and 29-times larger spinules without clathrin. Together, these findings suggest that the presence of a spinule is a marker for stronger and more stable presynaptic boutons in CA1, and that synaptic spinules serve at least two separable and distinct functions.
Collapse
|
60
|
Kozlova A, Zhang S, Kotlar AV, Jamison B, Zhang H, Shi S, Forrest MP, McDaid J, Cutler DJ, Epstein MP, Zwick ME, Pang ZP, Sanders AR, Warren ST, Gejman PV, Mulle JG, Duan J. Loss of function of OTUD7A in the schizophrenia- associated 15q13.3 deletion impairs synapse development and function in human neurons. Am J Hum Genet 2022; 109:1500-1519. [PMID: 35931052 PMCID: PMC9388388 DOI: 10.1016/j.ajhg.2022.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 06/27/2022] [Indexed: 02/06/2023] Open
Abstract
Identifying causative gene(s) within disease-associated large genomic regions of copy-number variants (CNVs) is challenging. Here, by targeted sequencing of genes within schizophrenia (SZ)-associated CNVs in 1,779 SZ cases and 1,418 controls, we identified three rare putative loss-of-function (LoF) mutations in OTU deubiquitinase 7A (OTUD7A) within the 15q13.3 deletion in cases but none in controls. To tie OTUD7A LoF with any SZ-relevant cellular phenotypes, we modeled the OTUD7A LoF mutation, rs757148409, in human induced pluripotent stem cell (hiPSC)-derived induced excitatory neurons (iNs) by CRISPR-Cas9 engineering. The mutant iNs showed a ∼50% decrease in OTUD7A expression without undergoing nonsense-mediated mRNA decay. The mutant iNs also exhibited marked reduction of dendritic complexity, density of synaptic proteins GluA1 and PSD-95, and neuronal network activity. Congruent with the neuronal phenotypes in mutant iNs, our transcriptomic analysis showed that the set of OTUD7A LoF-downregulated genes was enriched for those relating to synapse development and function and was associated with SZ and other neuropsychiatric disorders. These results suggest that OTUD7A LoF impairs synapse development and neuronal function in human neurons, providing mechanistic insight into the possible role of OTUD7A in driving neuropsychiatric phenotypes associated with the 15q13.3 deletion.
Collapse
Affiliation(s)
- Alena Kozlova
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | - Siwei Zhang
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA; Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA
| | - Alex V Kotlar
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA; Pillar Biosciences Inc., Natick, MA 01760, USA
| | - Brendan Jamison
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | - Hanwen Zhang
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | - Serena Shi
- Winston Churchill High School, Potomac, MD 20854, USA
| | - Marc P Forrest
- Department of Neuroscience, Northwestern University, Chicago, IL 60611, USA; Center for Autism and Neurodevelopment, Northwestern University, Chicago, IL 60611, USA
| | - John McDaid
- Department of Neurosurgery, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | - David J Cutler
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Michael P Epstein
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Michael E Zwick
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA; Senior Vice President for Research, Rutgers University, New Brunswick, NJ 08901, USA
| | - Zhiping P Pang
- Department of Neuroscience and Cell Biology, Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Alan R Sanders
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA; Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA
| | - Stephen T Warren
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Pablo V Gejman
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA; Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA
| | - Jennifer G Mulle
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Psychiatry, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ 08901, USA
| | - Jubao Duan
- Center for Psychiatric Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA; Department of Psychiatry and Behavioral Neuroscience, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
61
|
Synaptic plasticity during systems memory consolidation. Neurosci Res 2022; 183:1-6. [PMID: 35667493 DOI: 10.1016/j.neures.2022.05.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/29/2022] [Accepted: 05/31/2022] [Indexed: 11/23/2022]
Abstract
After learning, memory is initially encoded in the hippocampus but subsequently stabilized in other brain regions such as the cortex for long-lasting storage. This process is known as systems memory consolidation, and its cellular mechanism has long been a fundamental question. Synaptic plasticity is the major cellular mechanism underlying learning and memory, and is therefore considered a key function in the process of systems memory consolidation. Therefore, many studies have aimed to establish a causal link between synaptic plasticity in the brain and memory-associated behaviors. In this review, I discuss the various lines of research showing the function of synaptic plasticity, mainly in the hippocampus and cortex during memory consolidation.
Collapse
|
62
|
Yang Y, Liu JJ. Structural LTP: Signal transduction, actin cytoskeleton reorganization, and membrane remodeling of dendritic spines. Curr Opin Neurobiol 2022; 74:102534. [DOI: 10.1016/j.conb.2022.102534] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 02/14/2022] [Accepted: 03/03/2022] [Indexed: 01/05/2023]
|
63
|
Wang F, Cai YJ, Ma X, Wang N, Wu ZB, Sun Y, Xu YX, Yang H, Liu TT, Xia Q, Yu Z, Zhu DF. Synaptic loss in a mouse model of euthyroid Hashimoto's thyroiditis: possible involvement of the microglia. BMC Neurosci 2022; 23:25. [PMID: 35468730 PMCID: PMC9036731 DOI: 10.1186/s12868-022-00710-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 04/05/2022] [Indexed: 11/24/2022] Open
Abstract
Background Hashimoto’s thyroiditis (HT) is an autoimmune illness that renders individuals vulnerable to neuropsychopathology even in the euthyroid state, the mechanisms involved remain unclear. We hypothesized that activated microglia might disrupt synapses, resulting in cognitive disturbance in the context of euthyroid HT, and designed the present study to test this hypothesis. Methods Experimental HT model was induced by immunizing NOD mice with thyroglobulin and adjuvant twice. Morris Water Maze was measured to determine mice spatial learning and memory. The synaptic parameters such as the synaptic density, synaptic ultrastructure and synaptic-markers (SYN and PSD95) as well as the interactions of microglia with synapses were also determined. Results HT mice had poorer performance in Morris Water Maze than controls. Concurrently, HT resulted in a significant reduction in synapse density and ultrastructure damage, along with decreased synaptic puncta visualized by immunostaining with synaptophysin and PSD-95. In parallel, frontal activated microglia in euthyroid HT mice showed increased engulfment of PSD95 and EM revealed that the synaptic structures were visible within the microglia. These functional alterations in microglia corresponded to structural increases in their attachment to neuronal perikarya and a reduction in presynaptic terminals covering the neurons. Conclusion Our results provide initial evidence that HT can induce synaptic loss in the euthyroid state with deficits might be attributable to activated microglia, which may underlie the deleterious effects of HT on spatial learning and memory. Supplementary Information The online version contains supplementary material available at 10.1186/s12868-022-00710-2.
Collapse
Affiliation(s)
- Fen Wang
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Yao-Jun Cai
- Department of Endocrinology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Xiao Ma
- Department of Respiratoration, Wuhu Hospital of Traditional Chinese Medicine, Wuhu, 241000, China
| | - Nan Wang
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Zhang-Bi Wu
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Yan Sun
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Yong-Xia Xu
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Hao Yang
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Tian-Tian Liu
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Qin Xia
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Zhen Yu
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China.
| | - De-Fa Zhu
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
64
|
Stacho M, Manahan-Vaughan D. The Intriguing Contribution of Hippocampal Long-Term Depression to Spatial Learning and Long-Term Memory. Front Behav Neurosci 2022; 16:806356. [PMID: 35548697 PMCID: PMC9084281 DOI: 10.3389/fnbeh.2022.806356] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 03/10/2022] [Indexed: 01/03/2023] Open
Abstract
Long-term potentiation (LTP) and long-term depression (LTD) comprise the principal cellular mechanisms that fulfill established criteria for the physiological correlates of learning and memory. Traditionally LTP, that increases synaptic weights, has been ascribed a prominent role in learning and memory whereas LTD, that decreases them, has often been relegated to the category of "counterpart to LTP" that serves to prevent saturation of synapses. In contradiction of these assumptions, studies over the last several years have provided functional evidence for distinct roles of LTD in specific aspects of hippocampus-dependent associative learning and information encoding. Furthermore, evidence of the experience-dependent "pruning" of excitatory synapses, the majority of which are located on dendritic spines, by means of LTD has been provided. In addition, reports exist of the temporal and physical restriction of LTP in dendritic compartments by means of LTD. Here, we discuss the role of LTD and LTP in experience-dependent information encoding based on empirical evidence derived from conjoint behavioral and electrophysiological studies conducted in behaving rodents. We pinpoint the close interrelation between structural modifications of dendritic spines and the occurrence of LTP and LTD. We report on findings that support that whereas LTP serves to acquire the general scheme of a spatial representation, LTD enables retention of content details. We argue that LTD contributes to learning by engaging in a functional interplay with LTP, rather than serving as its simple counterpart, or negator. We propose that similar spatial experiences that share elements of neuronal representations can be modified by means of LTD to enable pattern separation. Therewith, LTD plays a crucial role in the disambiguation of similar spatial representations and the prevention of generalization.
Collapse
|
65
|
Wu XL, Yan QJ, Zhu F. Abnormal synaptic plasticity and impaired cognition in schizophrenia. World J Psychiatry 2022; 12:541-557. [PMID: 35582335 PMCID: PMC9048451 DOI: 10.5498/wjp.v12.i4.541] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 07/28/2021] [Accepted: 03/25/2022] [Indexed: 02/06/2023] Open
Abstract
Schizophrenia (SCZ) is a severe mental illness that affects several brain domains with relation to cognition and behaviour. SCZ symptoms are typically classified into three categories, namely, positive, negative, and cognitive. The etiology of SCZ is thought to be multifactorial and poorly understood. Accumulating evidence has indicated abnormal synaptic plasticity and cognitive impairments in SCZ. Synaptic plasticity is thought to be induced at appropriate synapses during memory formation and has a critical role in the cognitive symptoms of SCZ. Many factors, including synaptic structure changes, aberrant expression of plasticity-related genes, and abnormal synaptic transmission, may influence synaptic plasticity and play vital roles in SCZ. In this article, we briefly summarize the morphology of the synapse, the neurobiology of synaptic plasticity, and the role of synaptic plasticity, and review potential mechanisms underlying abnormal synaptic plasticity in SCZ. These abnormalities involve dendritic spines, postsynaptic density, and long-term potentiation-like plasticity. We also focus on cognitive dysfunction, which reflects impaired connectivity in SCZ. Additionally, the potential targets for the treatment of SCZ are discussed in this article. Therefore, understanding abnormal synaptic plasticity and impaired cognition in SCZ has an essential role in drug therapy.
Collapse
Affiliation(s)
- Xiu-Lin Wu
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Department of Medical Microbiology, School of Medicine, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Qiu-Jin Yan
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Department of Medical Microbiology, School of Medicine, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Fan Zhu
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Department of Medical Microbiology, School of Medicine, Wuhan University, Wuhan 430071, Hubei Province, China
| |
Collapse
|
66
|
Liu D, Lv F, Min S, Yang Y, Chen L. Inhibition of NLRP3 inflammasome-mediated neuroinflammation alleviates electroconvulsive shock-induced memory impairment via regulation of hippocampal synaptic plasticity in depressive rats. Behav Brain Res 2022; 428:113879. [PMID: 35390431 DOI: 10.1016/j.bbr.2022.113879] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/11/2022] [Accepted: 04/01/2022] [Indexed: 12/27/2022]
Abstract
Electroconvulsive shock has been considered one of the most effective treatment modalities for major depressive disorder. The association of acute transitory neuroinflammation in the hippocampus following electroconvulsive therapy with transient learning and memory impairment limits its clinical application. Whereas the NLRP3 inflammatory pathway is deemed to serve a key role in neuroinflammatory regulation, we aimed to examine if NLRP3 inflammasome activation was linked to electroconvulsive shock (ECS)-induced neuroinflammation and cognitive deficits. The depressed rats were modeled with chronic unpredictable mild stress. Their depression-like behaviors and cognitive performance were evaluated via sucrose preference test, forced swim test, open field test, and Morris water maze test. The NLRP3 expression was determined by western blot. The hippocampal CA1 region was immunohistochemically and electron-microscopically examined, respectively, for the activation of Iba-1 positive microglia and the ultrastructure of synapses. In this work, we found that ECS induced microglial activation in the rat hippocampal CA1 region. Pharmacological inhibition of NLRP3 inflammasome with MCC950 (NLRP3 inhibitor) in vivo significantly alleviated ECS-induced spatial learning and memory impairment, partially reversed neuroinflammation, and synaptic structural plasticity in the damaged hippocampal CA1 region, and reduced synapse associated protein expression and microglial activation. It offers a potential new approach for the prevention and treatment of cognitive decline following electroconvulsive therapy.
Collapse
Affiliation(s)
- Di Liu
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Feng Lv
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Su Min
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - You Yang
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lihao Chen
- Department of Anesthesiology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
67
|
Wang Y, Yang Y, Liu Y, Guo A, Zhang Y. Cognitive impairments in type 1 diabetes mellitus model mice are associated with synaptic protein disorders. Neurosci Lett 2022; 777:136587. [PMID: 35337951 DOI: 10.1016/j.neulet.2022.136587] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 01/24/2022] [Accepted: 03/21/2022] [Indexed: 11/24/2022]
Abstract
An association between type 1 diabetes mellitus (T1DM) and cognitive impairment was recently reported. However, the mechanisms by which T1DM induces cognitive impairment are still unknown. Here, we confirmed that T1DM mice induced by streptozotocin (STZ) injection had impaired working memory and spatial memory. We observed long-term potentiation (LTP) induction defects and synaptic loss in mice 20 weeks after STZ injection. We also found decreased levels of synaptic proteins, including the N-methyl-D-aspartic acid receptor (NMDAR) subunit NR2A, synaptophysin (SYP), and postsynaptic density 95 (PSD95), in the hippocampus and prefrontal cortex, revealing similarities in the alteration patterns of these synaptic proteins in aged Alzheimer's disease (AD) APP/PS1 mice and T1DM mice. Taken together, these findings expand our understanding of the mechanisms underlying T1DM-induced cognitive impairment.
Collapse
Affiliation(s)
- Yiming Wang
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Yueqi Yang
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Yiqiong Liu
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China
| | - Angyang Guo
- Duke Kunshan University, Kunshan, Jiangsu 215316, China
| | - Yan Zhang
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China.
| |
Collapse
|
68
|
Peng D, Qing X, Guan L, Li HY, Qiao L, Chen YB, Cai YF, Wang Q, Zhang SJ. Carnosine improves cognitive impairment through promoting SIRT6 expression and inhibiting ER stress in a diabetic encephalopathy model. Rejuvenation Res 2022; 25:79-88. [PMID: 35302398 DOI: 10.1089/rej.2022.0002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Diabetic encephalopathy is one of complications of diabetes mellitus. Carnosine is a dipeptide composed of β-alanine and L-histidine. Study has shown that carnosine could ameliorate cognitive impairment in animal model with diabetes mellitus. However, the mechanism remains unclear. An animal model of type 2 diabetes (db/db mice) was used in this study. The animals were treated with 0.9 % saline or carnosine (100 mg/kg) for 8 weeks. Morris water maze was tested after drug administration. Oxidative stress-related factors malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-PX), and pro-inflammatory factors inducible nitric oxide synthase (iNOS) were measured. Synapse-related protein postsynapticdensity 95 (PSD95) and brain-derived neurotrophic factor (BDNF) were detected by western blot. Besides, the expressions of sirtuin 6 (SIRT6), binding immunoglobulin protein (BIP), protein kinase R-like endoplasmic reticulum kinase (PERK), phospho-protein kinase R-like endoplasmic reticulum kinase (P-PERK), inositol-requiring enzyme-1α (IRE1α), phospho-inositol-requiring enzyme-1α (P-IRE1α), activating transcription factor 6 (ATF6), C/EBP-homologous protein (CHOP) in the hippocampus of the brain were detected. The results showed that treatment with carnosine ameliorated cognitive impairment in db/db mice. Carnosine reduced neuronal oxidative stress damage and iNOS expression in db/db mice. Meanwhile, carnosine relieved neurodegeneration in the hippocampus of db/db mice. Furthermore, carnosine promoted the expression of SIRT6 and reduced the expressions of endoplasmic reticulum (ER) related factors (BIP, P-PERK, P-IRE1α, ATF6, CHOP). In conclusion, these data suggested that the protective effect of carnosine against diabetic encephalopathy might be related to SIRT6/ER stress pathway.
Collapse
Affiliation(s)
- Dong Peng
- Guangzhou University of Chinese Medicine, 47879, Guangzhou, Guangdong, China;
| | - Xia Qing
- Guangzhou University of Chinese Medicine, 47879, Guangzhou, Guangdong, China;
| | - Li Guan
- Guangzhou University of Chinese Medicine, 47879, Guangzhou, China;
| | - Hong-Ying Li
- Guangzhou University of Chinese Medicine, 47879, Guangzhou, Guangdong, China;
| | - Lijun Qiao
- Guangzhou University of Chinese Medicine, 47879, Guangzhou, Guangdong, China;
| | - Yun-Bo Chen
- Guangzhou University of Chinese Medicine, 47879, Guangzhou, Guangdong, China;
| | - Ye-Feng Cai
- Guangzhou University of Chinese Medicine, 47879, Guangzhou, Guangdong, China;
| | - Qi Wang
- Guangzhou University of Chinese Medicine, 47879, Guangzhou, China;
| | - Shi-Jie Zhang
- Guangzhou University of Chinese Medicine, 47879, Guangzhou University of Chinese Medicine, Guangzhou, China, 510006;
| |
Collapse
|
69
|
Wegner W, Steffens H, Gregor C, Wolf F, Willig KI. Environmental enrichment enhances patterning and remodeling of synaptic nanoarchitecture as revealed by STED nanoscopy. eLife 2022; 11:73603. [PMID: 35195066 PMCID: PMC8903838 DOI: 10.7554/elife.73603] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 02/22/2022] [Indexed: 12/04/2022] Open
Abstract
Synaptic plasticity underlies long-lasting structural and functional changes to brain circuitry and its experience-dependent remodeling can be fundamentally enhanced by environmental enrichment. It is however unknown, whether and how the environmental enrichment alters the morphology and dynamics of individual synapses. Here, we present a virtually crosstalk-free two-color in vivo stimulated emission depletion (STED) microscope to simultaneously superresolve the dynamics of endogenous PSD95 of the post-synaptic density and spine geometry in the mouse cortex. In general, the spine head geometry and PSD95 assemblies were highly dynamic, their changes depended linearly on their original size but correlated only mildly. With environmental enrichment, the size distributions of PSD95 and spine head sizes were sharper than in controls, indicating that synaptic strength is set more uniformly. The topography of the PSD95 nanoorganization was more dynamic after environmental enrichment; changes in size were smaller but more correlated than in mice housed in standard cages. Thus, two-color in vivo time-lapse imaging of synaptic nanoorganization uncovers a unique synaptic nanoplasticity associated with the enhanced learning capabilities under environmental enrichment.
Collapse
Affiliation(s)
- Waja Wegner
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Göttingen, Germany
| | - Heinz Steffens
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Göttingen, Germany
| | - Carola Gregor
- Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Fred Wolf
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
| | - Katrin I Willig
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
70
|
Kylies J, Brunne B, Rune GM. A culture model for the assessment of phenylalanine neurotoxicity in phenylketonuria. IN VITRO MODELS 2022; 1:103-114. [PMID: 39872976 PMCID: PMC11756468 DOI: 10.1007/s44164-021-00007-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 11/09/2021] [Accepted: 11/16/2021] [Indexed: 01/30/2025]
Abstract
Objective Phenylketonuria (PKU) is caused by a specific mutation of the phenylalanine hydroxylase (PAH) gene. The deficiency of PAH results in high phenylalanine levels (Phe), low tyrosine levels (Tyr), and reduced catecholamine neurotransmitters. The majority of PKU patients, if untreated, develop severe mental retardation. The specific contribution of high Phe and low Tyr levels in mental retardation is largely unknown. In this study, we used organic hippocampal slice cultures in an optimized medium as an adequate culture model to decipher the precise role of high Phe and low Tyr levels on synaptic and glial integrity in PKU. The hippocampus is closely related to learning and memory and reduced catecholamine neurotransmitter levels can be neglected since these neurotransmitters do not derive from the hippocampus. Cultures exposed to physiological concentrations of Phe were compared with cultures exposed to doses of Phe/Tyr, as in the cerebral fluid of PKU patients. Methods Using capillary western blot analysis and immunohistochemistry, followed by quantitative image analysis, we tested the expression of various pre- and postsynaptic proteins (PSD95, synaptopodin, SNAP25, synaptophysin), glial cell markers (GFAP, Iba1, P2Y12, CD68, C3b), and the morphology of glial cells. Results We found a downregulation of the postsynaptic protein PSD95 and the presynaptic protein SNAP25 in the presence of high/low Phe/Tyr levels after 3 weeks, which, then however, recovered after 6 weeks in culture. Furthermore, no change in the expression pattern of glial proteins was observed. Conclusion Our results show that high Phe levels/low Tyr levels alone are unlikely to substantially contribute to mental retardation in PKU. The direct neurotoxic potency of high Phe/low Tyr concentrations is almost negligible since the effects are transient. The transient character in the presence of unchanged levels of high Phe/low Tyr points to a role of reduced catecholamine derivate neurotransmitters, rather than of high Phe/low Tyr levels in PKU.
Collapse
Affiliation(s)
- Julian Kylies
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Bianka Brunne
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Gabriele M. Rune
- Institute of Cell Biology and Neurobiology, Charité Anatomy, Charité Universitätsmedizin Berlin, Charitéplatz 1 (intern: Virchowweg 6 CCO), 10117 Berlin, Germany
| |
Collapse
|
71
|
2D Ti 3C 2T xMXene couples electrical stimulation to promote proliferation and neural differentiation of neural stem cells. Acta Biomater 2022; 139:105-117. [PMID: 33348061 DOI: 10.1016/j.actbio.2020.12.035] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 01/31/2023]
Abstract
Preclinical studies involving stem cells require efficient physiochemical regulations on the fate of such cells. Because of their unique planar structure, metallic conductivity, and flexible surface functionalization, MXenes show potential for modulating stem cell fate. Here, the Ti3C2TxMXenenanosheets are dispersed on tissue culture polystyrene (TCPS). When primary mouse neural stem cells (NSCs) are cultured on laminin-coated Ti3C2TxMXene film, they form stable adhesion, retain their proliferative ability, and show extensive spreading of terminal extensions. With respect to their functional activity, NSCs cultured on Ti3C2TxMXene films form more active and synchronous network activity than those cultured on TCPS substrates. Moreover, Ti3C2TxMXene film significantly promotes the neural differentiation and the neurons have longer neurites and greater numbers of branch points and branch tips. NSC-derived neurons grown on the Ti3C2Tx MXene film preserved normal synapse development. Finally, electrical stimulation coupled with Ti3C2TxMXene film significantly enhances the proliferation of NSCs. These results indicate that Ti3C2TxMXene is an efficient interface for the proliferation and neural differentiation of NSC and the maturation of NSC-derived neurons, which expands the potential uses of the MXene family of materials and provides new strategies for stem cell studies. STATEMENT OF SIGNIFICANCE: The 2DTi3C2TxMXenenanosheets were applied to be an interface for regulating neural stem cells (NSCs). NSCs cultured on Ti3C2TxMXene film possessed higher proliferative ability with higher and more synchronous electrical activities. Moreover, Ti3C2TxMXene film significantly promoted the neural differentiation ratio of NSCs, and the neurons derived from NSCs cultured on Ti3C2TxMXene film had longer neurites and greater numbers of branch points and branch tips.When electrical stimulation was applied to NSCs via the Ti3C2TxMXene film, it significantly enhanced the proliferation of NSCs. This work expands the potential uses of the MXene family of materials and provides new strategies for stem cell studies.
Collapse
|
72
|
Sodium hydrosulfide reverses β 2-microglobulin-induced depressive-like behaviors of male Sprague-Dawley rats: Involving improvement of synaptic plasticity and enhancement of Warburg effect in hippocampus. Behav Brain Res 2022; 417:113562. [PMID: 34499939 DOI: 10.1016/j.bbr.2021.113562] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 08/03/2021] [Accepted: 08/27/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Our previous works demonstrated that β2-microglobulin (β2m), a systemic pro-aging factor, induce depressive-like behaviors. Hydrogen sulfide (H2S) is identified as a potential target for treatment of depression. The aim of the present work is to explore whether H2S antagonizes β2m-induced depressive-like behaviors and the underlying mechanisms. METHODS The depressive-like behaviors were detected using the novelty suppressed feeding test (NSFT), tail suspension test (TST), forced swimming test (FST) and open field test (OFT). The expressions of Warburg-related proteins, including hexokinase II (HK II), pyruvate kinase M2 (PKM2), Lactate dehydrogenase A (LDHA), pyruvate dehydrogenase (PDH) and pyruvate dehydrogenase kinase 1(PDK1), and synaptic plasticity-related proteins, including postsynaptic density protein 95 (PSD95) and synaptophysin1 (SYN1), were determined by western blotting. RESULT we found that NaHS (the donor of H2S) attenuated the depressive-like behaviors in the β2m-exposed rats, as judged by NSFT, TST, FST, and OFT. We also demonstrated that NaHS enhanced the synaptic plasticity, as evidenced by the upregulations of PSD95 and SYN1 expressions in the hippocampus of β2m-exposed rats. Furthermore, NaHS improved the Warburg effect in the hippocampus of β2m-exposed rats, as evidenced by the upregulations of HK II, PKM2, LDHA and PDK1 expressions, and the downregulation of PDH expression. CONCLUSION H2S prevents β2m-induced depressive-like behaviors, which is involved in improvement of hippocampal synaptic plasticity as a result of enhancement of hippocampal Warburg effect.
Collapse
|
73
|
MEK1/2 inhibition rescues neurodegeneration by TFEB-mediated activation of autophagic lysosomal function in a model of Alzheimer's Disease. Mol Psychiatry 2022; 27:4770-4780. [PMID: 35948663 PMCID: PMC9734062 DOI: 10.1038/s41380-022-01713-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 07/10/2022] [Accepted: 07/15/2022] [Indexed: 12/14/2022]
Abstract
Alzheimer's Disease (AD) is a progressive neurodegenerative disorder, which is characterized by cognitive deficit due to synaptic loss and neuronal death. Extracellular amyloid β plaques are one of the pathological hallmarks of AD. The autophagic lysosomal pathway is the essential mechanism to maintain cellular homeostasis by driving clearance of protein aggregates and is dysfunctional in AD. Here, we showed that inhibiting MEK/ERK signaling using a clinically available MEK1/2 inhibitor, trametinib (GSK1120212, SNR1611), induces the protection of neurons through autophagic lysosomal activation mediated by transcription factor EB (TFEB) in a model of AD. Orally administered trametinib recovered impaired neural structures, cognitive functions, and hippocampal long-term potentiation (LTP) in 5XFAD mice. Trametinib also reduced Aβ deposition via induction of autophagic lysosomal activation. RNA-sequencing analysis revealed upregulation of autophagic lysosomal genes by trametinib administration. In addition, trametinib inhibited TFEB phosphorylation at Ser142 and promoted its nuclear translocation, which in turn induced autophagic lysosomal related genes, indicating that trametinib activates the autophagic lysosomal process through TFEB activation. From these observations, we concluded that MEK inhibition provides neuronal protection from the Aβ burden by increasing autophagic lysosomal activity. Thus, MEK inhibition may be an effective therapeutic strategy for AD.
Collapse
|
74
|
Hosokawa T, Liu PW. Regulation of the Stability and Localization of Post-synaptic Membrane Proteins by Liquid-Liquid Phase Separation. Front Physiol 2021; 12:795757. [PMID: 34975543 PMCID: PMC8716852 DOI: 10.3389/fphys.2021.795757] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 11/17/2021] [Indexed: 12/01/2022] Open
Abstract
Synaptic plasticity is a cellular mechanism of learning and memory. The synaptic strength can be persistently upregulated or downregulated to update the information sent to the neuronal network and form a memory engram. For its molecular mechanism, the stability of α-amino-3-hydroxyl-5-methyl-4-isoxazolepropionate-type glutamate receptor (AMPAR), a glutamatergic ionotropic receptor, on the postsynaptic membrane has been studied for these two decades. Since AMPAR is not saturated on the postsynaptic membrane during a single event of neurotransmitter release, the number and nanoscale localization of AMPAR is critical for regulating the efficacy of synaptic transmission. The observation of AMPAR on the postsynaptic membrane by super-resolution microscopy revealed that AMPAR forms a nanodomain that is defined as a stable segregated cluster on the postsynaptic membrane to increase the efficacy of synaptic transmission. Postsynaptic density (PSD), an intracellular protein condensate underneath the postsynaptic membrane, regulates AMPAR dynamics via the intracellular domain of Stargazin, an auxiliary subunit of AMPAR. Recently, it was reported that PSD is organized by liquid-liquid phase separation (LLPS) to form liquid-like protein condensates. Furthermore, the calcium signal induced by the learning event triggers the persistent formation of sub-compartments of different protein groups inside protein condensates. This explains the formation of nanodomains via synaptic activation. The liquid-like properties of LLPS protein condensates are ideal for the molecular mechanism of synaptic plasticity. In this review, we summarize the recent progress in the properties and regulation of synaptic plasticity, postsynaptic receptors, PSD, and LLPS.
Collapse
|
75
|
Vallés AS, Barrantes FJ. Nanoscale Sub-Compartmentalization of the Dendritic Spine Compartment. Biomolecules 2021; 11:1697. [PMID: 34827695 PMCID: PMC8615865 DOI: 10.3390/biom11111697] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 01/04/2023] Open
Abstract
Compartmentalization of the membrane is essential for cells to perform highly specific tasks and spatially constrained biochemical functions in topographically defined areas. These membrane lateral heterogeneities range from nanoscopic dimensions, often involving only a few molecular constituents, to micron-sized mesoscopic domains resulting from the coalescence of nanodomains. Short-lived domains lasting for a few milliseconds coexist with more stable platforms lasting from minutes to days. This panoply of lateral domains subserves the great variety of demands of cell physiology, particularly high for those implicated in signaling. The dendritic spine, a subcellular structure of neurons at the receiving (postsynaptic) end of central nervous system excitatory synapses, exploits this compartmentalization principle. In its most frequent adult morphology, the mushroom-shaped spine harbors neurotransmitter receptors, enzymes, and scaffolding proteins tightly packed in a volume of a few femtoliters. In addition to constituting a mesoscopic lateral heterogeneity of the dendritic arborization, the dendritic spine postsynaptic membrane is further compartmentalized into spatially delimited nanodomains that execute separate functions in the synapse. This review discusses the functional relevance of compartmentalization and nanodomain organization in synaptic transmission and plasticity and exemplifies the importance of this parcelization in various neurotransmitter signaling systems operating at dendritic spines, using two fast ligand-gated ionotropic receptors, the nicotinic acetylcholine receptor and the glutamatergic receptor, and a second-messenger G-protein coupled receptor, the cannabinoid receptor, as paradigmatic examples.
Collapse
Affiliation(s)
- Ana Sofía Vallés
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (UNS-CONICET), Bahía Blanca 8000, Argentina;
| | - Francisco J. Barrantes
- Laboratory of Molecular Neurobiology, Institute of Biomedical Research (BIOMED), UCA-CONICET, Av. Alicia Moreau de Justo 1600, Buenos Aires C1107AFF, Argentina
| |
Collapse
|
76
|
Zhang H, Su Y, Sun Z, Chen M, Han Y, Li Y, Dong X, Ding S, Fang Z, Li W, Li W. Ginsenoside Rg1 alleviates Aβ deposition by inhibiting NADPH oxidase 2 activation in APP/PS1 mice. J Ginseng Res 2021; 45:665-675. [PMID: 34764721 PMCID: PMC8569324 DOI: 10.1016/j.jgr.2021.03.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 02/25/2021] [Accepted: 03/10/2021] [Indexed: 01/18/2023] Open
Abstract
Background Ginsenoside Rg1 (Rg1), an active ingredient in ginseng, may be a potential agent for the treatment of Alzheimer’s disease (AD). However, the protective effect of Rg1 on neurodegeneration in AD and its mechanism of action are still incompletely understood. Methods Wild type (WT) and APP/PS1 AD mice, from 6 to 9 months old, were used in the experiment. The open field test (OFT) and Morris water maze (MWM) were used to detect behavioral changes. Neuronal damage was assessed by hematoxylin and eosin (H&E) and Nissl staining. Immunofluorescence, western blotting, and quantitative real-time polymerase chain reaction (q-PCR) were used to examine postsynaptic density 95 (PSD95) expression, amyloid beta (Aβ) deposition, Tau and phosphorylated Tau (p-Tau) expression, reactive oxygen species (ROS) production, and NAPDH oxidase 2 (NOX2) expression. Results Rg1 treatment for 12 weeks significantly ameliorated cognitive impairments and neuronal damage and decreased the p-Tau level, amyloid precursor protein (APP) expression, and Aβ generation in APP/PS1 mice. Meanwhile, Rg1 treatment significantly decreased the ROS level and NOX2 expression in the hippocampus and cortex of APP/PS1 mice. Conclusions Rg1 alleviates cognitive impairments, neuronal damage, and reduce Aβ deposition by inhibiting NOX2 activation in APP/PS1 mice. Rg1 treatment significantly alleviated cognitive dysfunction and neuronal damage in APP/PS1 mice. Rg1 treatment significantly reduced APP expression and Aβ deposition in APP/PS1 mice. The expression of NOX2 and ROS production were significantly increased in APP/PS1 mice. Rg1 treatment significantly decreased NOX2 expression and ROS accumulation in APP/PS1 mice.
Collapse
Affiliation(s)
- Han Zhang
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei, China
| | - Yong Su
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhenghao Sun
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei, China
| | - Ming Chen
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei, China
| | - Yuli Han
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei, China
| | - Yan Li
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei, China
| | - Xianan Dong
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei, China
| | - Shixin Ding
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei, China
| | - Zhirui Fang
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei, China
| | - Weiping Li
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei, China
| | - Weizu Li
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei, China
| |
Collapse
|
77
|
Silent Synapses in Cocaine-Associated Memory and Beyond. J Neurosci 2021; 41:9275-9285. [PMID: 34759051 DOI: 10.1523/jneurosci.1559-21.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/22/2021] [Accepted: 09/27/2021] [Indexed: 11/21/2022] Open
Abstract
Glutamatergic synapses are key cellular sites where cocaine experience creates memory traces that subsequently promote cocaine craving and seeking. In addition to making across-the-board synaptic adaptations, cocaine experience also generates a discrete population of new synapses that selectively encode cocaine memories. These new synapses are glutamatergic synapses that lack functionally stable AMPARs, often referred to as AMPAR-silent synapses or, simply, silent synapses. They are generated de novo in the NAc by cocaine experience. After drug withdrawal, some of these synapses mature by recruiting AMPARs, contributing to the consolidation of cocaine-associated memory. After cue-induced retrieval of cocaine memories, matured silent synapses alternate between two dynamic states (AMPAR-absent vs AMPAR-containing) that correspond with the behavioral manifestations of destabilization and reconsolidation of these memories. Here, we review the molecular mechanisms underlying silent synapse dynamics during behavior, discuss their contributions to circuit remodeling, and analyze their role in cocaine-memory-driven behaviors. We also propose several mechanisms through which silent synapses can form neuronal ensembles as well as cross-region circuit engrams for cocaine-specific behaviors. These perspectives lead to our hypothesis that cocaine-generated silent synapses stand as a distinct set of synaptic substrates encoding key aspects of cocaine memory that drive cocaine relapse.
Collapse
|
78
|
Lenz M, Eichler A, Kruse P, Muellerleile J, Deller T, Jedlicka P, Vlachos A. All-trans retinoic acid induces synaptopodin-dependent metaplasticity in mouse dentate granule cells. eLife 2021; 10:71983. [PMID: 34723795 PMCID: PMC8560091 DOI: 10.7554/elife.71983] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/15/2021] [Indexed: 12/27/2022] Open
Abstract
Previously we showed that the vitamin A metabolite all-trans retinoic acid (atRA) induces synaptic plasticity in acute brain slices prepared from the mouse and human neocortex (Lenz et al., 2021). Depending on the brain region studied, distinct effects of atRA on excitatory and inhibitory neurotransmission have been reported. Here, we used intraperitoneal injections of atRA (10 mg/kg) in adult C57BL/6J mice to study the effects of atRA on excitatory and inhibitory neurotransmission in the mouse fascia dentata—a brain region implicated in memory acquisition. No major changes in synaptic transmission were observed in the ventral hippocampus while a significant increase in both spontaneous excitatory postsynaptic current frequencies and synapse numbers were evident in the dorsal hippocampus 6 hr after atRA administration. The intrinsic properties of hippocampal dentate granule cells were not significantly different and hippocampal transcriptome analysis revealed no essential neuronal changes upon atRA treatment. In light of these findings, we tested for the metaplastic effects of atRA, that is, for its ability to modulate synaptic plasticity expression in the absence of major changes in baseline synaptic strength. Indeed, in vivo long-term potentiation (LTP) experiments demonstrated that systemic atRA treatment improves the ability of dentate granule cells to express LTP. The plasticity-promoting effects of atRA were not observed in synaptopodin-deficient mice, therefore, extending our previous results regarding the relevance of synaptopodin in atRA-mediated synaptic strengthening in the mouse prefrontal cortex. Taken together, our data show that atRA mediates synaptopodin-dependent metaplasticity in mouse dentate granule cells.
Collapse
Affiliation(s)
- Maximilian Lenz
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Amelie Eichler
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Pia Kruse
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julia Muellerleile
- Institute of Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Neuroscience Center, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Neuroscience Center, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Peter Jedlicka
- Institute of Clinical Neuroanatomy, Dr. Senckenberg Anatomy, Neuroscience Center, Goethe-University Frankfurt, Frankfurt am Main, Germany.,ICAR3R - Interdisciplinary Centre for 3Rs in Animal Research, Faculty of Medicine, Justus-Liebig-University, Giessen, Germany
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Basics in Neuromodulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center Brain Links Brain Tools, University of Freiburg, Freiburg, Germany
| |
Collapse
|
79
|
Díaz-Alonso J, Nicoll RA. AMPA receptor trafficking and LTP: Carboxy-termini, amino-termini and TARPs. Neuropharmacology 2021; 197:108710. [PMID: 34271016 PMCID: PMC9122021 DOI: 10.1016/j.neuropharm.2021.108710] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/28/2021] [Accepted: 07/08/2021] [Indexed: 12/11/2022]
Abstract
AMPA receptors (AMPARs) are fundamental elements in excitatory synaptic transmission and synaptic plasticity in the CNS. Long term potentiation (LTP), a form of synaptic plasticity which contributes to learning and memory formation, relies on the accumulation of AMPARs at the postsynapse. This phenomenon requires the coordinated recruitment of different elements in the AMPAR complex. Based on recent research reviewed herein, we propose an updated AMPAR trafficking and LTP model which incorporates both extracellular as well as intracellular mechanisms. This article is part of the special Issue on 'Glutamate Receptors - AMPA receptors'.
Collapse
Affiliation(s)
- Javier Díaz-Alonso
- Department of Anatomy and Neurobiology, USA; Center for the Neurobiology of Learning and Memory, University of California at Irvine, USA.
| | - Roger A Nicoll
- Departments of Cellular and Molecular Pharmacology, USA; Physiology, University of California at San Francisco, USA.
| |
Collapse
|
80
|
Kudryashova IV. The Reorganization of the Actin Matrix as a Factor of Presynaptic Plasticity. NEUROCHEM J+ 2021. [DOI: 10.1134/s1819712421030089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
81
|
Sun Y, Smirnov M, Kamasawa N, Yasuda R. Rapid Ultrastructural Changes in the PSD and Surrounding Membrane after Induction of Structural LTP in Single Dendritic Spines. J Neurosci 2021; 41:7003-7014. [PMID: 34266899 PMCID: PMC8372018 DOI: 10.1523/jneurosci.1964-20.2021] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 06/05/2021] [Accepted: 06/10/2021] [Indexed: 11/21/2022] Open
Abstract
The structural plasticity of dendritic spines is considered to be an important basis of synaptic plasticity, learning, and memory. Here, we induced input-specific structural LTP (sLTP) in single dendritic spines in organotypic hippocampal slices from mice of either sex and performed ultrastructural analyses of the spines using efficient correlative light and electron microscopy. We observed reorganization of the PSD nanostructure, such as perforation and segmentation, at 2-3, 20, and 120 min after sLTP induction. In addition, PSD and nonsynaptic axon-spine interface (nsASI) membrane expanded unevenly during sLTP. Specifically, the PSD area showed a transient increase at 2-3 min after sLTP induction. The PSD growth was to a degree less than spine volume growth at 2-3 min and 20 min after sLTP induction but became similar at 120 min. On the other hand, the nsASI area showed a profound and lasting expansion, to a degree similar to spine volume growth throughout the process. These rapid ultrastructural changes in PSD and surrounding membrane may contribute to rapid electrophysiological plasticity during sLTP.SIGNIFICANCE STATEMENT To understand the ultrastructural changes during synaptic plasticity, it is desired to efficiently image single dendritic spines that underwent structural plasticity in electron microscopy. We induced structural long-term potentiation (sLTP) in single dendritic spines by two-photon glutamate uncaging. We then identified the same spines at different phases of sLTP and performed ultrastructural analysis by using an efficient correlative light and electron microscopy method. We found that postsynaptic density undergoes dramatic modification in its structural complexity immediately after sLTP induction. Meanwhile, the nonsynaptic axon-spine interface area shows a rapid and sustained increase throughout sLTP. Our results indicate that the uneven modification of synaptic and nonsynaptic postsynaptic membrane might contribute to rapid electrophysiological plasticity during sLTP.
Collapse
Affiliation(s)
- Ye Sun
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458
- Integrative Biology and Neuroscience Graduate Program, Florida Atlantic University, Jupiter, Florida 33458
- International Max Planck Research School for Brain and Behavior, Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458
| | - Michael Smirnov
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458
| | - Naomi Kamasawa
- Electron Microscopy Core Facility, Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458
- International Max Planck Research School for Brain and Behavior, Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458
| | - Ryohei Yasuda
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458
- Integrative Biology and Neuroscience Graduate Program, Florida Atlantic University, Jupiter, Florida 33458
- International Max Planck Research School for Brain and Behavior, Max Planck Florida Institute for Neuroscience, Jupiter, Florida 33458
| |
Collapse
|
82
|
Ma D, Liu J, Wei C, Shen W, Yang Y, Lin D, Wu A. Activation of CD200-CD200R1 Axis Attenuates Perioperative Neurocognitive Disorder Through Inhibition of Neuroinflammation in Mice. Neurochem Res 2021; 46:3190-3199. [PMID: 34392443 DOI: 10.1007/s11064-021-03422-x] [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: 11/09/2020] [Revised: 07/24/2021] [Accepted: 08/04/2021] [Indexed: 10/20/2022]
Abstract
Perioperative neurocognitive disorder (PND) is the mild cognitive impairment associated with surgery and anesthesia. It is a common surgical complication in the elderly. An important mechanism of PND is the surgically induced neuroinflammation. The interaction between the neuronal surface protein CD200 and its receptor in microglia, CD200R1, is an important regulatory pathway to control neuroinflammation. However, the potential role of the CD200-CD200R1 pathway in the acute period of PND has not been fully investigated. In this study, in a PND mouse model, we first measured the protein expression level of CD200, CD200R1, and the related pro- and anti-inflammatory cytokines in the hippocampus. Then, we investigated cognitive function, neuroinflammation and postsynaptic density protein 95 (PSD-95) expression after the injection of CD200-Fc (agonist), CD200R1-Fc (antagonist) or IgG1-Fc (vehicle) into lateral ventricle in PND models. Compared with the control group, the expression of CD200 was up-regulated at day 1 after surgery in PND models. The injection of the CD200-Fc into the lateral ventricle could mitigate primed neuroinflammation and cognitive decline, increase the expression of PSD-95 at day 1 after surgery in PND models. In conclusion, we have demonstrated that CD200-CD200R1 signaling was involved in the acute inflammatory process of PND, and activating CD200R1 can inhibit neuroinflammation and attenuate PND. Thus, the CD200-CD200R1 axis is a potential novel target for PND prevention and treatment.
Collapse
Affiliation(s)
- Danxu Ma
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, No. 8 Gongtinan Road, Chaoyang District, Beijing, 100020, People's Republic of China
| | - Jinhu Liu
- Department of Anesthesiology, Beijing First Hospital of Integrated Traditional Chinese and Western Medicine, Beijing, 100021, People's Republic of China
| | - Changwei Wei
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, No. 8 Gongtinan Road, Chaoyang District, Beijing, 100020, People's Republic of China
| | - Wenzhen Shen
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, No. 8 Gongtinan Road, Chaoyang District, Beijing, 100020, People's Republic of China
| | - Yinan Yang
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, No. 8 Gongtinan Road, Chaoyang District, Beijing, 100020, People's Republic of China
| | - Dandan Lin
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, No. 8 Gongtinan Road, Chaoyang District, Beijing, 100020, People's Republic of China
| | - Anshi Wu
- Department of Anesthesiology, Beijing Chaoyang Hospital, Capital Medical University, No. 8 Gongtinan Road, Chaoyang District, Beijing, 100020, People's Republic of China.
| |
Collapse
|
83
|
Hayashi Y. Molecular mechanism of hippocampal long-term potentiation - Towards multiscale understanding of learning and memory. Neurosci Res 2021; 175:3-15. [PMID: 34375719 DOI: 10.1016/j.neures.2021.08.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 11/16/2022]
Abstract
Long-term potentiation (LTP) of synaptic transmission is considered to be a cellular counterpart of learning and memory. Activation of postsynaptic NMDA type glutamate receptor (NMDA-R) induces trafficking of AMPA type glutamate receptors (AMPA-R) and other proteins to the synapse in sequential fashion. At the same time, the dendritic spine expands for long-term and modulation of actin underlies this (structural LTP or sLTP). How these changes persist despite constant diffusion and turnover of the component proteins have been the central focus of the current LTP research. Signaling triggered by Ca2+-influx via NMDA-R triggers kinase including Ca2+/calmodulin-dependent protein kinase II (CaMKII). CaMKII can sustain longer-term biochemical signaling by forming a reciprocally-activating kinase-effector complex with its substrate proteins including Tiam1, thereby regulating persistence of the downstream signaling. Furthermore, activated CaMKII can condense at the synapse through the mechanism of liquid-liquid phase separation (LLPS). This increases the binding capacity at the synapse, thereby contributing to the maintenance of enlarged protein complexes. It may also serve as the synapse tag, which captures newly synthesized proteins.
Collapse
Affiliation(s)
- Yasunori Hayashi
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan.
| |
Collapse
|
84
|
Alizadeh Makvandi A, Khalili M, Roghani M, Amiri Moghaddam S. Hesperetin ameliorates electroconvulsive therapy-induced memory impairment through regulation of hippocampal BDNF and oxidative stress in a rat model of depression. J Chem Neuroanat 2021; 117:102001. [PMID: 34246766 DOI: 10.1016/j.jchemneu.2021.102001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 06/14/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022]
Abstract
Depression is one of the most common mental health disorders and it is generally characterized by negative mood. Although electroconvulsive therapy (ECT) is an effective treatment for depression, however, it can cause cognitive deficit. Hesperetin, an active ingredient in citrus peels, has antioxidant and neuroprotective properties. In this study, we evaluated the effect of hesperetin on memory impairment induced by ECT in a reserpine-induced depression model in male rat. For this purpose, 105 male rats weighing 230-250 g were randomly divided into control and reserpine-treated groups. The reserpine-treated animals were subdivided into: Reserpine, Hesperetin (10 and 20 mg/kg), ECT and ECT+Hesperetin (10 and 20 mg/kg). After taking the drugs, the effect of hesperetin was evaluated through behavioral NORT, Y Maze, FST, SPT and also via assessment of hippocampal brain-derived neurotrophic factor (BDNF) and oxidative stress biomarkers i.e., MDA, SOD and GSH. As a result, our biochemical studies showed a significant decrease of MDA in groups treated with ECT+Hesperetin as compared to ECT and hesperetin groups (P < 0.001) and a marked increase in SOD, GSH and BDNF in ECT+Hesperetin 20 group as compared to other groups (p < 0.05). Also, the results of behavioral tests revealed that treatment with hesperetin can increase the novel object recognition index and alternation behaviors in Y maze test as compared to the groups treated with hesperetin or ECT (p < 0.05). These results suggest that co-administration of hesperetin with ECT is effective for improvement of cognitive function and can alleviate ECT-induced memory impairment in reserpine-treated rats.
Collapse
Affiliation(s)
| | - Mohsen Khalili
- Neurophysiology Research Center, Shahed University, Tehran, Iran.
| | - Mehrdad Roghani
- Neurophysiology Research Center, Shahed University, Tehran, Iran.
| | - Sara Amiri Moghaddam
- Department of Pharmacy, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
85
|
Hu Y, Li D, Wei H, Zhou S, Chen W, Yan X, Cai J, Chen X, Chen B, Liao M, Chai R, Tang M. Neurite Extension and Orientation of Spiral Ganglion Neurons Can Be Directed by Superparamagnetic Iron Oxide Nanoparticles in a Magnetic Field. Int J Nanomedicine 2021; 16:4515-4526. [PMID: 34239302 PMCID: PMC8259836 DOI: 10.2147/ijn.s313673] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/21/2021] [Indexed: 12/19/2022] Open
Abstract
Introduction Neuroregeneration is a major challenge in neuroscience for treating degenerative diseases and for repairing injured nerves. Numerous studies have shown the importance of physical stimulation for neuronal growth and development, and here we report an approach for the physical guidance of neuron orientation and neurite growth using superparamagnetic iron oxide (SPIO) nanoparticles and magnetic fields (MFs). Methods SPIO nanoparticles were synthesized by classic chemical co-precipitation methods and then characterized by transmission electron microscope, dynamic light scattering, and vibrating sample magnetometer. The cytotoxicity of the prepared SPIO nanoparticles and MF was determined using CCK-8 assay and LIVE/DEAD assay. The immunofluorescence images were captured by a laser scanning confocal microscopy. Cell migration was evaluated using the wound healing assay. Results The prepared SPIO nanoparticles showed a narrow size distribution, low cytotoxicity, and superparamagnetism. SPIO nanoparticles coated with poly-L-lysine could be internalized by spiral ganglion neurons (SGNs) and showed no cytotoxicity at concentrations less than 300 µg/mL. The neurite extension of SGNs was promoted after internalizing SPIO nanoparticles with or without an external MF, and this might be due to the promotion of growth cone development. It was also confirmed that SPIO can regulate cell migration and can direct neurite outgrowth in SGNs preferentially along the direction imposed by an external MF. Conclusion Our results provide a fundamental understanding of the regulation of cell behaviors under physical cues and suggest alternative treatments for sensorineural hearing loss caused by the degeneration of SGNs.
Collapse
Affiliation(s)
- Yangnan Hu
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, People's Republic of China
| | - Dan Li
- School of Biology, Food and Environment, Hefei University, Hefei, 230601, People's Republic of China
| | - Hao Wei
- Department of Otorhinolaryngology Head and Neck Surgery, Drum Tower Clinical Medical College, Nanjing Medical University, Nanjing, 210000, People's Republic of China
| | - Shan Zhou
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, People's Republic of China
| | - Wei Chen
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, People's Republic of China
| | - Xiaoqian Yan
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, People's Republic of China
| | - Jaiying Cai
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, People's Republic of China
| | - Xiaoyan Chen
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, People's Republic of China
| | - Bo Chen
- Materials Science and Devices Institute, Suzhou University of Science and Technology, Suzhou, 215009, People's Republic of China
| | - Menghui Liao
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, People's Republic of China
| | - Renjie Chai
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, People's Republic of China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, People's Republic of China
| | - Mingliang Tang
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, 210096, People's Republic of China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, People's Republic of China.,Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Medical College, Soochow University, Suzhou, 215000, People's Republic of China
| |
Collapse
|
86
|
Zhang LQ, Zhang W, Li T, Yang T, Yuan X, Zhou Y, Zou Q, Yang H, Gao F, Tian Y, Mei W, Tian XB. GLP-1R activation ameliorated novel-object recognition memory dysfunction via regulating hippocampal AMPK/NF-κB pathway in neuropathic pain mice. Neurobiol Learn Mem 2021; 182:107463. [PMID: 34015440 DOI: 10.1016/j.nlm.2021.107463] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 05/02/2021] [Accepted: 05/12/2021] [Indexed: 01/15/2023]
Abstract
Growing evidences indicate that neuropathic pain is frequently accompanied with cognitive impairments, which aggravate the decrease in the quality of life of chronic pain patients. Furthermore, it has been shown that the activation of Glucagon-like-peptide-1receptor (GLP-1R) improved memory deficit in multiple diseases, including Alzheimer's disease (AD), stroke. However, whether GLP-1R activation could improve memory impairment induced by neuropathic pain and the mechanisms underlying the effect of the activation of GLP-1R on memory protection have not yet been established. The spared nerve injury (SNI) model was established as a kind of neuropathic pain. And novel-object recognition memory (hippocampus-dependent memory) was tested by the novel object recognition test (NORT). The expression levels of GLP-1, GLP-1R, adenosine monophosphate-activated protein kinase (AMPK), p-AMPKThr172, nuclear factor κ B p65 (NF-κB p65), interleukin-1beta (IL-1β), IL-1β p17 (mature IL-1β), tumor necrosis factor-alpha (TNF-α) and the synaptic proteins were tested in the murine hippocampus with memory deficits caused by neuropathic pain. Then, exenatide acetate (Ex-4, a GLP-1R agonist), exendin (9-39) (Ex(9-39), a GLP-1R antagonist) and Compound C dihydrochloride (CC, an AMPK inhibitor) were used to test the effects of the activation of GLP-1R in the mice with neuropathic pain. First, we uncovered that neuropathic pain could inhibit GLP-1/GLP-R axis, disturb inflammatory signaling pathway, increase the expression of IL-1β, IL-1β p17 and TNF-α, downregulate the synaptic proteins (postsynaptic density protein 95 (PSD95) and Arc). Subsequently, we reported that Ex-4 treatment could improve recognition memory impairment, increase the ratio of p-AMPKThr172/AMPK, inhibit the phosphorylation NF-κB p65 and decrease the expression of IL-1β, IL-1β p17 and TNF-α, upregulate the levels of PSD95 and Arc. Moreover, we found that Ex(9-39) and CC treatment could abrogate the memory protection of activation of GLP-1R in mice with neuropathic pain. The results indicated that the activation of GLP-1R could improve recognition memory impairment via regulating AMPK/NF-κB pathway, improving neuroinflammation, reversing the decreased level of synaptic proteins in neuropathic pain mice.
Collapse
Affiliation(s)
- Long-Qing Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wen Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ting Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ting Yang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoman Yuan
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yaqun Zhou
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qian Zou
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hui Yang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Feng Gao
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - YuKe Tian
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Mei
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xue-Bi Tian
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
87
|
Gennaccaro L, Fuchs C, Loi M, Pizzo R, Alvente S, Berteotti C, Lupori L, Sagona G, Galvani G, Gurgone A, Raspanti A, Medici G, Tassinari M, Trazzi S, Ren E, Rimondini R, Pizzorusso T, Giovanna Z, Maurizio G, Elisabetta C. Age-Related Cognitive and Motor Decline in a Mouse Model of CDKL5 Deficiency Disorder is Associated with Increased Neuronal Senescence and Death. Aging Dis 2021; 12:764-785. [PMID: 34094641 PMCID: PMC8139207 DOI: 10.14336/ad.2020.0827] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 08/27/2020] [Indexed: 01/02/2023] Open
Abstract
CDKL5 deficiency disorder (CDD) is a severe neurodevelopmental disease caused by mutations in the X-linked CDKL5 gene. Children affected by CDD display a clinical phenotype characterized by early-onset epilepsy, intellectual disability, motor impairment, and autistic-like features. Although the clinical aspects associated with CDKL5 mutations are well described in children, adults with CDD are still under-characterized. Similarly, most animal research has been carried out on young adult Cdkl5 knockout (KO) mice only. Since age represents a risk factor for the worsening of symptoms in many neurodevelopmental disorders, understanding age differences in the development of behavioral deficits is crucial in order to optimize the impact of therapeutic interventions. Here, we compared young adult Cdkl5 KO mice with middle-aged Cdkl5 KO mice, at a behavioral, neuroanatomical, and molecular level. We found an age-dependent decline in motor, cognitive, and social behaviors in Cdkl5 KO mice, as well as in breathing and sleep patterns. The behavioral decline in older Cdkl5 KO mice was not associated with a worsening of neuroanatomical alterations, such as decreased dendritic arborization or spine density, but was paralleled by decreased neuronal survival in different brain regions such as the hippocampus, cortex, and basal ganglia. Interestingly, we found increased β-galactosidase activity and DNA repair protein levels, γH2AX and XRCC5, in the brains of older Cdkl5 KO mice, which suggests that an absence of Cdkl5 accelerates neuronal senescence/death by triggering irreparable DNA damage. In summary, this work provides evidence that CDKL5 may play a fundamental role in neuronal survival during brain aging and suggests a possible worsening with age of the clinical picture in CDD patients.
Collapse
Affiliation(s)
- Laura Gennaccaro
- 1Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Claudia Fuchs
- 1Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Manuela Loi
- 1Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Riccardo Pizzo
- 2Department of Neuroscience, University of Turin, Turin, Italy
| | - Sara Alvente
- 1Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Chiara Berteotti
- 1Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Leonardo Lupori
- 3BIO@SNS lab, Scuola Normale Superiore di Pisa, Pisa, Italy.,4Institute of Neuroscience, National Research Council, Pisa, Italy
| | - Giulia Sagona
- 4Institute of Neuroscience, National Research Council, Pisa, Italy.,5Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA, University of Florence, Florence, Italy.,6Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, Pisa, Italy
| | - Giuseppe Galvani
- 1Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Antonia Gurgone
- 2Department of Neuroscience, University of Turin, Turin, Italy
| | | | - Giorgio Medici
- 1Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Marianna Tassinari
- 1Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Stefania Trazzi
- 1Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Elisa Ren
- 1Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Roberto Rimondini
- 7Department of Medical and Clinical Sciences, University of Bologna, Bologna, Italy
| | - Tommaso Pizzorusso
- 3BIO@SNS lab, Scuola Normale Superiore di Pisa, Pisa, Italy.,4Institute of Neuroscience, National Research Council, Pisa, Italy.,5Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA, University of Florence, Florence, Italy
| | - Zoccoli Giovanna
- 1Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Giustetto Maurizio
- 2Department of Neuroscience, University of Turin, Turin, Italy.,8National Institute of Neuroscience-Italy, Turin, Italy
| | - Ciani Elisabetta
- 1Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
88
|
Feng Z, Wu X, Zhang M. Presynaptic bouton compartmentalization and postsynaptic density-mediated glutamate receptor clustering via phase separation. Neuropharmacology 2021; 193:108622. [PMID: 34051266 DOI: 10.1016/j.neuropharm.2021.108622] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/25/2021] [Accepted: 05/17/2021] [Indexed: 01/21/2023]
Abstract
Neuronal synapses encompass three compartments: presynaptic axon terminal, synaptic cleft, and postsynaptic dendrite. Each compartment contains densely packed molecular machineries that are involved in synaptic transmission. In recent years, emerging evidence indicates that the assembly of these membraneless substructures or assemblies that are not enclosed by membranes are driven by liquid-liquid phase separation. We review here recent studies that suggest the phase separation-mediated organization of these synaptic compartments. We discuss how synaptic function may be linked to its organization as biomolecular condensates. We conclude with a discussion of areas of future interest in the field for better understanding of the structural architecture of neuronal synapses and its contribution to synaptic functions.
Collapse
Affiliation(s)
- Zhe Feng
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Institute for Advanced Study, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Xiandeng Wu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Mingjie Zhang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
89
|
The importance of ultrastructural analysis of memory. Brain Res Bull 2021; 173:28-36. [PMID: 33984429 DOI: 10.1016/j.brainresbull.2021.04.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/19/2021] [Accepted: 04/22/2021] [Indexed: 11/22/2022]
Abstract
Plasticity of glutamatergic synapses in the hippocampus is believed to underlie learning and memory processes. Surprisingly, very few studies report long-lasting structural changes of synapses induced by behavioral training. It remains, therefore, unclear which synaptic changes in the hippocampus contribute to memory storage. Here, we systematically compare how long-term potentiation of synaptic transmission (LTP) (a primary form of synaptic plasticity and cellular model of memory) and behavioral training affect hippocampal glutamatergic synapses at the ultrastructural level enabled by electron microscopy. The review of the literature indicates that while LTP induces growth of dendritic spines and post-synaptic densities (PSD), that represent postsynaptic part of a glutamatergic synapse, after behavioral training there is transient (< 6 h) synaptogenesis and long-lasting (> 24 h) increase in PSD volume (without a significant change of dendritic spine volume), indicating that training-induced PSD growth may reflect long-term enhancement of synaptic functions. Additionally, formation of multi-innervated spines (MIS), is associated with long-term memory in aged mice and LTP-deficient mutant mice. Since volume of PSD, as well as atypical synapses, can be reliably observed only with electron microscopy, we argue that the ultrastructural level of analysis is required to reveal synaptic changes that are associated with long-term storage of information in the brain.
Collapse
|
90
|
Pérez-Sisqués L, Martín-Flores N, Masana M, Solana-Balaguer J, Llobet A, Romaní-Aumedes J, Canal M, Campoy-Campos G, García-García E, Sánchez-Fernández N, Fernández-García S, Gilbert JP, Rodríguez MJ, Man HY, Feinstein E, Williamson DL, Soto D, Gasull X, Alberch J, Malagelada C. RTP801 regulates motor cortex synaptic transmission and learning. Exp Neurol 2021; 342:113755. [PMID: 33984337 DOI: 10.1016/j.expneurol.2021.113755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/24/2021] [Accepted: 05/08/2021] [Indexed: 01/26/2023]
Abstract
BACKGROUND RTP801/REDD1 is a stress-regulated protein whose upregulation is necessary and sufficient to trigger neuronal death in in vitro and in vivo models of Parkinson's and Huntington's diseases and is up regulated in compromised neurons in human postmortem brains of both neurodegenerative disorders. Indeed, in both Parkinson's and Huntington's disease mouse models, RTP801 knockdown alleviates motor-learning deficits. RESULTS We investigated the physiological role of RTP801 in neuronal plasticity and we found RTP801 in rat, mouse and human synapses. The absence of RTP801 enhanced excitatory synaptic transmission in both neuronal cultures and brain slices from RTP801 knock-out (KO) mice. Indeed, RTP801 KO mice showed improved motor learning, which correlated with lower spine density but increased basal filopodia and mushroom spines in the motor cortex layer V. This paralleled with higher levels of synaptosomal GluA1 and TrkB receptors in homogenates derived from KO mice motor cortex, proteins that are associated with synaptic strengthening. CONCLUSIONS Altogether, these results indicate that RTP801 has an important role modulating neuronal plasticity and motor learning. They will help to understand its role in neurodegenerative disorders where RTP801 levels are detrimentally upregulated.
Collapse
Affiliation(s)
- Leticia Pérez-Sisqués
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain.
| | - Núria Martín-Flores
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain
| | - Mercè Masana
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain; IDIBAPS- Institut d'Investigacions BiomèdiquesAugust Pi i Sunyer, Barcelona, 08036, Catalonia, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, 08036, Catalonia, Spain
| | - Júlia Solana-Balaguer
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain
| | - Arnau Llobet
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain
| | - Joan Romaní-Aumedes
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain
| | - Mercè Canal
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain
| | - Genís Campoy-Campos
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain
| | - Esther García-García
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain; IDIBAPS- Institut d'Investigacions BiomèdiquesAugust Pi i Sunyer, Barcelona, 08036, Catalonia, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, 08036, Catalonia, Spain
| | | | - Sara Fernández-García
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain; IDIBAPS- Institut d'Investigacions BiomèdiquesAugust Pi i Sunyer, Barcelona, 08036, Catalonia, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, 08036, Catalonia, Spain
| | - James P Gilbert
- Department of Biology, Pharmacology and Experimental Therapeutics, Boston University, Boston, MA 02215, USA
| | - Manuel José Rodríguez
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain; IDIBAPS- Institut d'Investigacions BiomèdiquesAugust Pi i Sunyer, Barcelona, 08036, Catalonia, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, 08036, Catalonia, Spain
| | - Heng-Ye Man
- Department of Biology, Pharmacology and Experimental Therapeutics, Boston University, Boston, MA 02215, USA
| | | | - David L Williamson
- Kinesiology Program, School of Behavioral Sciences and Education, Penn State Harrisburg, Middletown, PA 17057, USA
| | - David Soto
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain; IDIBAPS- Institut d'Investigacions BiomèdiquesAugust Pi i Sunyer, Barcelona, 08036, Catalonia, Spain
| | - Xavier Gasull
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain; IDIBAPS- Institut d'Investigacions BiomèdiquesAugust Pi i Sunyer, Barcelona, 08036, Catalonia, Spain
| | - Jordi Alberch
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain; IDIBAPS- Institut d'Investigacions BiomèdiquesAugust Pi i Sunyer, Barcelona, 08036, Catalonia, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, 08036, Catalonia, Spain
| | - Cristina Malagelada
- Department of Biomedicine, Faculty of Medicine, University of Barcelona, Catalonia, Spain; Institut de Neurociències, University of Barcelona, 08036, Catalonia, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, 08036, Catalonia, Spain.
| |
Collapse
|
91
|
Holderith N, Heredi J, Kis V, Nusser Z. A High-Resolution Method for Quantitative Molecular Analysis of Functionally Characterized Individual Synapses. Cell Rep 2021; 32:107968. [PMID: 32726631 PMCID: PMC7408500 DOI: 10.1016/j.celrep.2020.107968] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/15/2020] [Accepted: 07/08/2020] [Indexed: 12/24/2022] Open
Abstract
Elucidating the molecular mechanisms underlying the functional diversity of synapses requires a high-resolution, sensitive, diffusion-free, quantitative localization method that allows the determination of many proteins in functionally characterized individual synapses. Array tomography permits the quantitative analysis of single synapses but has limited sensitivity, and its application to functionally characterized synapses is challenging. Here, we aim to overcome these limitations by searching the parameter space of different fixation, resin, embedding, etching, retrieval, and elution conditions. Our optimizations reveal that etching epoxy-resin-embedded ultrathin sections with Na-ethanolate and treating them with SDS dramatically increase the labeling efficiency of synaptic proteins. We also demonstrate that this method is ideal for the molecular characterization of individual synapses following paired recordings, two-photon [Ca2+] or glutamate-sensor (iGluSnFR) imaging. This method fills a missing gap in the toolbox of molecular and cellular neuroscience, helping us to reveal how molecular heterogeneity leads to diversity in function. Etching and antigen retrieval enhance immunoreactions in epoxy-resin-embedded tissue Biocytin-filled nerve cells can be visualized in epoxy-resin-embedded tissue Molecular composition of functionally characterized individual synapses is revealed Multiplexed, postembedding reactions are compatible with STED imaging
Collapse
Affiliation(s)
- Noemi Holderith
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Judit Heredi
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Viktor Kis
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Budapest 1083, Hungary
| | - Zoltan Nusser
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Budapest 1083, Hungary.
| |
Collapse
|
92
|
Karlocai MR, Heredi J, Benedek T, Holderith N, Lorincz A, Nusser Z. Variability in the Munc13-1 content of excitatory release sites. eLife 2021; 10:67468. [PMID: 33904397 PMCID: PMC8116053 DOI: 10.7554/elife.67468] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 04/26/2021] [Indexed: 01/15/2023] Open
Abstract
The molecular mechanisms underlying the diversity of cortical glutamatergic synapses are still incompletely understood. Here, we tested the hypothesis that presynaptic active zones (AZs) are constructed from molecularly uniform, independent release sites (RSs), the number of which scales linearly with the AZ size. Paired recordings between hippocampal CA1 pyramidal cells and fast-spiking interneurons in acute slices from adult mice followed by quantal analysis demonstrate large variability in the number of RSs (N) at these connections. High-resolution molecular analysis of functionally characterized synapses reveals variability in the content of one of the key vesicle priming factors – Munc13-1 – in AZs that possess the same N. Replica immunolabeling also shows a threefold variability in the total Munc13-1 content of AZs of identical size and a fourfold variability in the size and density of Munc13-1 clusters within the AZs. Our results provide evidence for quantitative molecular heterogeneity of RSs and support a model in which the AZ is built up from variable numbers of molecularly heterogeneous, but independent RSs.
Collapse
Affiliation(s)
- Maria Rita Karlocai
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Judit Heredi
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Tünde Benedek
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Noemi Holderith
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Andrea Lorincz
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Zoltan Nusser
- Laboratory of Cellular Neurophysiology, Institute of Experimental Medicine, Budapest, Hungary
| |
Collapse
|
93
|
Misquitta KA, Miles A, Prevot TD, Knoch JK, Fee C, Newton DF, Ellegood J, Lerch JP, Sibille E, Nikolova YS, Banasr M. Reduced anterior cingulate cortex volume induced by chronic stress correlates with increased behavioral emotionality and decreased synaptic puncta density. Neuropharmacology 2021; 190:108562. [PMID: 33864799 DOI: 10.1016/j.neuropharm.2021.108562] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 10/21/2022]
Abstract
Clinical and preclinical studies report that chronic stress induces behavioral deficits as well as volumetric and synaptic alterations in corticolimbic brain regions including the anterior cingulate cortex (ACC), amygdala (AMY), nucleus accumbens (NAc) and hippocampus (HPC). Here, we aimed to investigate the volumetric changes associated with chronic restraint stress (CRS) and link these changes to the CRS-induced behavioral and synaptic deficits. We first confirmed that CRS increases behavioral emotionality, defined as collective scoring of anxiety- and anhedonia-like behaviors. We then demonstrated that CRS induced a reduction of total brain volume which negatively correlated with behavioral emotionality. Region-specific analysis identified that only the ACC showed significant decrease in volume following CRS (p < 0.05). Reduced ACC correlated with increased behavioral emotionality (r = -0.56; p = 0.0003). Although not significantly altered by CRS, AMY and NAc (but not the HPC) volumes were negatively correlated with behavioral emotionality. Finally, using structural covariance network analysis to assess shared volumetric variances between the corticolimbic brain regions and associated structures, we found a progressive decreased ACC degree and increased AMY degree following CRS. At the cellular level, reduced ACC volume correlated with decreased PSD95 (but not VGLUT1) puncta density (r = 0.35, p < 0.05), which also correlated with increased behavioral emotionality (r = -0.44, p < 0.01), suggesting that altered synaptic strength is an underlying substrate of CRS volumetric and behavioral effects. Our results demonstrate that CRS effects on ACC volume and synaptic density are linked to behavioral emotionality and highlight key ACC structural and morphological alterations relevant to stress-related illnesses including mood and anxiety disorders.
Collapse
Affiliation(s)
- Keith A Misquitta
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada; Departments of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Amy Miles
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada
| | - Thomas D Prevot
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Jaime K Knoch
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada; Departments of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Corey Fee
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada; Departments of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Dwight F Newton
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada; Departments of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Jacob Ellegood
- Mouse Imaging Centre (MICe), Hospital for Sick Children, Toronto, Canada
| | - Jason P Lerch
- Mouse Imaging Centre (MICe), Hospital for Sick Children, Toronto, Canada; Wellcome Centre for Integrative Neuroimaging, FMRIB, Nuffield Department of Clinical Neuroscience, The University of Oxford, Oxford, UK; Department of Medical Biophysics, The University of Toronto, Toronto, Canada
| | - Etienne Sibille
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada; Departments of Pharmacology and Toxicology, University of Toronto, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Yuliya S Nikolova
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Mounira Banasr
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health (CAMH), Toronto, Canada; Departments of Pharmacology and Toxicology, University of Toronto, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada.
| |
Collapse
|
94
|
Obashi K, Taraska JW, Okabe S. The role of molecular diffusion within dendritic spines in synaptic function. J Gen Physiol 2021; 153:e202012814. [PMID: 33720306 PMCID: PMC7967910 DOI: 10.1085/jgp.202012814] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/16/2021] [Indexed: 12/21/2022] Open
Abstract
Spines are tiny nanoscale protrusions from dendrites of neurons. In the cortex and hippocampus, most of the excitatory postsynaptic sites reside in spines. The bulbous spine head is connected to the dendritic shaft by a thin membranous neck. Because the neck is narrow, spine heads are thought to function as biochemically independent signaling compartments. Thus, dynamic changes in the composition, distribution, mobility, conformations, and signaling properties of molecules contained within spines can account for much of the molecular basis of postsynaptic function and regulation. A major factor in controlling these changes is the diffusional properties of proteins within this small compartment. Advances in measurement techniques using fluorescence microscopy now make it possible to measure molecular diffusion within single dendritic spines directly. Here, we review the regulatory mechanisms of diffusion in spines by local intra-spine architecture and discuss their implications for neuronal signaling and synaptic plasticity.
Collapse
Affiliation(s)
- Kazuki Obashi
- Biochemistry and Biophysics Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Justin W. Taraska
- Biochemistry and Biophysics Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Shigeo Okabe
- Department of Cellular Neurobiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
95
|
Rulan D, Zhenbang Y, Yipu Z, Yuan G, Galaj E, Xiaorui S, Wenshuya L, Jiaqi L, Yan Z, Chang Y, Xi Y, Li S, Yixiao L, Haishui S. Exogenous SO 2 donor treatment impairs reconsolidation of drug reward memory in mice. Eur J Pharmacol 2021; 896:173911. [PMID: 33503460 DOI: 10.1016/j.ejphar.2021.173911] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/16/2021] [Accepted: 01/21/2021] [Indexed: 12/25/2022]
Abstract
Substance-related and addictive disorders (SRADs) are characterized by compulsive drug use and recurrent relapse. The persistence of pathological drug-related memories indisputably contributes to a high propensity to relapse. Hence, strategies to disrupt reconsolidation of drug reward memory are currently being pursued as potential anti-relapse interventions. Sulfur dioxide (SO2), acting as a potential gaseous molecule, endogenously derives from sulfur amino acid and can exert significant neural regulatory effects. However, the role of SO2 in reconsolidation of drug memory has not been determined. In the present study, we used morphine- or cocaine-induced conditioned place preference (CPP) mouse models with retrieval to investigate the effects of exogenous SO2 donor treatment on reconsolidation of drug reward memory. We found that administration of SO2 donor immediately after the retrieval impaired the expression of morphine or cocaine CPP. Furthermore, the exogenous SO2 donor treatment 6 h post-retrieval or in the absence of retrieval had no effect on drug reward memory and the expression of CPP. SO2 itself did not produce aversive effects nor did it acutely block morphine CPP. Our results indicate that exogenous SO2 impairs reconsolidation of drug reward memory rather than inhibits the expression of drug reward memory. As such, SO2 holds potential for the treatment and prevention of SRADs and should be studied further.
Collapse
Affiliation(s)
- Ding Rulan
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410081, China; Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yang Zhenbang
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Zhang Yipu
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Gao Yuan
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Ewa Galaj
- National Institute on Drug Abuse, Molecular Targets and Medications Discovery Branch, Baltimore, MD, USA
| | - Shi Xiaorui
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Li Wenshuya
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Luo Jiaqi
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Zhang Yan
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Yang Chang
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410081, China
| | - Yin Xi
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410081, China; Department of Functional Region of Diagnosis, Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Song Li
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China
| | - Luo Yixiao
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410081, China.
| | - Shi Haishui
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, 050017, China; Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medicinal University, 050017, China.
| |
Collapse
|
96
|
Ramos-Fernández E, Arrázola MS, Oliva CA, Arredondo SB, Varela-Nallar L, Inestrosa NC. Wnt5a promotes hippocampal postsynaptic development and GluN2B-induced expression via the eIF2α HRI kinase. Sci Rep 2021; 11:7395. [PMID: 33795747 PMCID: PMC8016897 DOI: 10.1038/s41598-021-86708-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/18/2021] [Indexed: 12/30/2022] Open
Abstract
Wnt signaling plays a key role in neurodevelopment and neuronal maturation. Specifically, Wnt5a stimulates postsynaptic assemblies, increases glutamatergic neurotransmission and, through calcium signaling, generates nitric oxide (NO). Trying to unveil the molecular pathway triggering these postsynaptic effects, we found that Wnt5a treatment induces a time-dependent increases in the length of the postsynaptic density (PSD), elicits novel synaptic contacts and facilitates F-actin flow both in in vitro and ex vivo models. These effects were partially abolished by the inhibition of the Heme-regulated eukaryotic initiation factor 2α (HRI) kinase, a kinase which phosphorylates the initiation translational factor eIF2α. When phosphorylated, eIF2α normally avoids the translation of proteins not needed during stress conditions, in order to avoid unnecessary energetic expenses. However, phosphorylated eIF2α promotes the translation of some proteins with more than one open reading frame in its 5′ untranslated region. One of these proteins targeted by Wnt-HRI-eIF2α mediated translation is the GluN2B subunit of the NMDA receptor. The identified increase in GluN2B expression correlated with increased NMDA receptor function. Considering that NMDA receptors are crucial for excitatory synaptic transmission, the molecular pathway described here contributes to the understanding of the fast and plastic translational mechanisms activated during learning and memory processes.
Collapse
Affiliation(s)
- Eva Ramos-Fernández
- Centro de Envejecimiento y Regeneración (CARE UC), CARE UC Biomedical Center, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Alameda 340, 8331150, Santiago, Chile. .,École polytechnique fédérale de Lausanne, Lausanne, Switzerland.
| | - Macarena S Arrázola
- Centro de Envejecimiento y Regeneración (CARE UC), CARE UC Biomedical Center, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Alameda 340, 8331150, Santiago, Chile.,Centro de Biología Integrativa, Universidad Mayor, Santiago, Chile
| | - Carolina A Oliva
- Centro de Envejecimiento y Regeneración (CARE UC), CARE UC Biomedical Center, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Alameda 340, 8331150, Santiago, Chile
| | - Sebastián B Arredondo
- Instituto de Ciencias Biomédicas, Facultad de Medicina y Facultad de Ciencias de La Vida, Universidad Andrés Bello, Santiago, Chile
| | - Lorena Varela-Nallar
- Instituto de Ciencias Biomédicas, Facultad de Medicina y Facultad de Ciencias de La Vida, Universidad Andrés Bello, Santiago, Chile
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE UC), CARE UC Biomedical Center, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Alameda 340, 8331150, Santiago, Chile. .,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile.
| |
Collapse
|
97
|
Bonilla-Quintana M, Wörgötter F. Exploring new roles for actin upon LTP induction in dendritic spines. Sci Rep 2021; 11:7072. [PMID: 33782451 PMCID: PMC8007616 DOI: 10.1038/s41598-021-86367-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 03/15/2021] [Indexed: 01/21/2023] Open
Abstract
Dendritic spines, small protrusions of the dendrites, enlarge upon LTP induction, linking morphological and functional properties. Although the role of actin in spine enlargement has been well studied, little is known about its relationship with mechanical membrane properties, such as membrane tension, which is involved in many cell processes, like exocytosis. Here, we use a 3D model of the dendritic spine to investigate how polymerization of actin filaments can effectively elevate the membrane tension to trigger exocytosis in a domain close to the tip of the spine. Moreover, we show that the same pool of actin promotes full membrane fusion after exocytosis and spine stabilization.
Collapse
|
98
|
Proietti Onori M, van Woerden GM. Role of calcium/calmodulin-dependent kinase 2 in neurodevelopmental disorders. Brain Res Bull 2021; 171:209-220. [PMID: 33774142 DOI: 10.1016/j.brainresbull.2021.03.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 01/28/2023]
Abstract
Neurodevelopmental disorders are a complex and heterogeneous group of neurological disorders characterized by their early-onset and estimated to affect more than 3% of children worldwide. The rapid advancement of sequencing technologies in the past years allowed the identification of hundreds of variants in several different genes causing neurodevelopmental disorders. Between those, new variants in the Calcium/calmodulin dependent protein kinase II (CAMK2) genes were recently linked to intellectual disability. Despite many years of research on CAMK2, this proves for the first time that this well-known and highly conserved molecule plays an important role in the human brain. In this review, we give an overview of the identified CAMK2 variants, and we speculate on potential mechanisms through which dysfunctions in CAMK2 result in neurodevelopmental disorders. Additionally, we discuss how the identification of CAMK2 variants might result in new exciting discoveries regarding the function of CAMK2 in the human brain.
Collapse
Affiliation(s)
- Martina Proietti Onori
- Department of Neuroscience, Erasmus MC, Rotterdam, 3015 GD, the Netherlands; The ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, 3015 GD, the Netherlands
| | - Geeske M van Woerden
- Department of Neuroscience, Erasmus MC, Rotterdam, 3015 GD, the Netherlands; The ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Rotterdam, 3015 GD, the Netherlands.
| |
Collapse
|
99
|
Carbone BE, Abouleish M, Watters KE, Vogel S, Ribic A, Schroeder OHU, Bader BM, Biederer T. Synaptic Connectivity and Cortical Maturation Are Promoted by the ω-3 Fatty Acid Docosahexaenoic Acid. Cereb Cortex 2021; 30:226-240. [PMID: 31034037 DOI: 10.1093/cercor/bhz083] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 02/20/2019] [Accepted: 03/27/2019] [Indexed: 12/14/2022] Open
Abstract
Brain development is likely impacted by micronutrients. This is supported by the effects of the ω-3 fatty acid docosahexaenoic acid (DHA) during early neuronal differentiation, when it increases neurite growth. Aiming to delineate DHA roles in postnatal stages, we selected the visual cortex due to its stereotypic maturation. Immunohistochemistry showed that young mice that received dietary DHA from birth exhibited more abundant presynaptic and postsynaptic specializations. DHA also increased density and size of synapses in a dose-dependent manner in cultured neurons. In addition, dendritic arbors of neurons treated with DHA were more complex. In agreement with improved connectivity, DHA enhanced physiological parameters of network maturation in vitro, including bursting strength and oscillatory behavior. Aiming to analyze functional maturation of the cortex, we performed in vivo electrophysiological recordings from awake mice to measure responses to patterned visual inputs. Dietary DHA robustly promoted the developmental increase in visual acuity, without altering light sensitivity. The visual acuity of DHA-supplemented animals continued to improve even after their cortex had matured and DHA abolished the acuity plateau. Our findings show that the ω-3 fatty acid DHA promotes synaptic connectivity and cortical processing. These results provide evidence that micronutrients can support the maturation of neuronal networks.
Collapse
Affiliation(s)
- Beatrice E Carbone
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Malik Abouleish
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Katherine E Watters
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Seth Vogel
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Adema Ribic
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | | | | | - Thomas Biederer
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
100
|
PSD-95 in CA1 Area Regulates Spatial Choice Depending on Age. J Neurosci 2021; 41:2329-2343. [PMID: 33472821 DOI: 10.1523/jneurosci.1996-20.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/06/2020] [Accepted: 12/21/2020] [Indexed: 11/21/2022] Open
Abstract
Cognitive processes that require spatial information rely on synaptic plasticity in the dorsal CA1 area (dCA1) of the hippocampus. Since the function of the hippocampus is impaired in aged individuals, it remains unknown how aged animals make spatial choices. Here, we used IntelliCage to study behavioral processes that support spatial choices of aged female mice living in a group. As a proxy of training-induced synaptic plasticity, we analyzed the morphology of dendritic spines and the expression of a synaptic scaffold protein, PSD-95. We observed that spatial choice training in young adult mice induced correlated shrinkage of dendritic spines and downregulation of PSD-95 in dCA1. Moreover, long-term depletion of PSD-95 by shRNA in dCA1 limited correct choices to a reward corner, while reward preference was intact. In contrast, old mice used behavioral strategies characterized by an increased tendency for perseverative visits and social interactions. This strategy resulted in a robust preference for the reward corner during the spatial choice task. Moreover, training decreased the correlation between PSD-95 expression and the size of dendritic spines. Furthermore, PSD-95 depletion did not impair place choice or reward preference in old mice. Thus, our data indicate that while young mice require PSD-95-dependent synaptic plasticity in dCA1 to make correct spatial choices, old animals observe cage mates and stick to a preferred corner to seek the reward. This strategy is resistant to the depletion of PSD-95 in the CA1 area. Overall, our study demonstrates that aged mice combine alternative behavioral and molecular strategies to approach and consume rewards in a complex environment.SIGNIFICANCE STATEMENT It remains poorly understood how aging affects behavioral and molecular processes that support cognitive functions. It is, however, essential to understand these processes to develop therapeutic interventions that support successful cognitive aging. Our data indicate that while young mice require PSD-95-dependent synaptic plasticity in dCA1 to make correct spatial choices (i.e., choices that require spatial information), old animals observe cage mates and stick to a preferred corner to seek the reward. This strategy is resistant to the depletion of PSD-95 in the CA1 area. Overall, our study demonstrates that aged mice combine alternative behavioral and molecular strategies to approach and consume rewards in a complex environment. Second, the contribution of PSD-95-dependent synaptic functions in spatial choice changes with age.
Collapse
|