1
|
Karpova A, Aly AAA, Marosi EL, Mikulovic S. Fiber-based in vivo imaging: unveiling avenues for exploring mechanisms of synaptic plasticity and neuronal adaptations underlying behavior. NEUROPHOTONICS 2024; 11:S11507. [PMID: 38390518 PMCID: PMC10883581 DOI: 10.1117/1.nph.11.s1.s11507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/24/2024]
Abstract
In recent decades, various subfields within neuroscience, spanning molecular, cellular, and systemic dimensions, have significantly advanced our understanding of the elaborate molecular and cellular mechanisms that underpin learning, memory, and adaptive behaviors. There have been notable advancements in imaging techniques, particularly in reaching superficial brain structures. This progress has led to their widespread adoption in numerous laboratories. However, essential physiological and cognitive processes, including sensory integration, emotional modulation of motivated behavior, motor regulation, learning, and memory consolidation, are intricately encoded within deeper brain structures. Hence, visualization techniques such as calcium imaging using miniscopes have gained popularity for studying brain activity in unrestrained animals. Despite its utility, miniscope technology is associated with substantial brain tissue damage caused by gradient refractive index lens implantation. Furthermore, its imaging capabilities are primarily confined to the neuronal somata level, thus constraining a comprehensive exploration of subcellular processes underlying adaptive behaviors. Consequently, the trajectory of neuroscience's future hinges on the development of minimally invasive optical fiber-based endo-microscopes optimized for cellular, subcellular, and molecular imaging within the intricate depths of the brain. In pursuit of this goal, select research groups have invested significant efforts in advancing this technology. In this review, we present a perspective on the potential impact of this innovation on various aspects of neuroscience, enabling the functional exploration of in vivo cellular and subcellular processes that underlie synaptic plasticity and the neuronal adaptations that govern behavior.
Collapse
Affiliation(s)
- Anna Karpova
- Leibniz Institute for Neurobiology, RG Neuroplasticity, Magdeburg, Germany
- Otto von Guericke University, Center for Behavioral Brain Sciences, Magdeburg, Germany
| | - Ahmed A. A. Aly
- Leibniz Institute for Neurobiology, RG Neuroplasticity, Magdeburg, Germany
| | - Endre Levente Marosi
- Leibniz Institute for Neurobiology, RG Cognition and Emotion, Magdeburg, Germany
| | - Sanja Mikulovic
- Otto von Guericke University, Center for Behavioral Brain Sciences, Magdeburg, Germany
- Leibniz Institute for Neurobiology, RG Cognition and Emotion, Magdeburg, Germany
- German Centre for Mental Health (DZPG), Magdeburg, Germany
| |
Collapse
|
2
|
Zecevic D. Electrical properties of dendritic spines. Biophys J 2023; 122:4303-4315. [PMID: 37837192 PMCID: PMC10698282 DOI: 10.1016/j.bpj.2023.10.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 09/27/2023] [Accepted: 10/11/2023] [Indexed: 10/15/2023] Open
Abstract
Dendritic spines are small protrusions that mediate most of the excitatory synaptic transmission in the brain. Initially, the anatomical structure of spines has suggested that they serve as isolated biochemical and electrical compartments. Indeed, following ample experimental evidence, it is now widely accepted that a significant physiological role of spines is to provide biochemical compartmentalization in signal integration and plasticity in the nervous system. In contrast to the clear biochemical role of spines, their electrical role is uncertain and is currently being debated. This is mainly because spines are small and not accessible to conventional experimental methods of electrophysiology. Here, I focus on reviewing the literature on the electrical properties of spines, including the initial morphological and theoretical modeling studies, indirect experimental approaches based on measurements of diffusional resistance of the spine neck, indirect experimental methods using two-photon uncaging of glutamate on spine synapses, optical imaging of intracellular calcium concentration changes, and voltage imaging with organic and genetically encoded voltage-sensitive probes. The interpretation of evidence from different preparations obtained with different methods has yet to reach a consensus, with some analyses rejecting and others supporting an electrical role of spines in regulating synaptic signaling. Thus, there is a need for a critical comparison of the advantages and limitations of different methodological approaches. The only experimental study on electrical signaling monitored optically with adequate sensitivity and spatiotemporal resolution using voltage-sensitive dyes concluded that mushroom spines on basal dendrites of cortical pyramidal neurons in brain slices have no electrical role.
Collapse
Affiliation(s)
- Dejan Zecevic
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut.
| |
Collapse
|
3
|
Kastellakis G, Tasciotti S, Pandi I, Poirazi P. The dendritic engram. Front Behav Neurosci 2023; 17:1212139. [PMID: 37576932 PMCID: PMC10412934 DOI: 10.3389/fnbeh.2023.1212139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/11/2023] [Indexed: 08/15/2023] Open
Abstract
Accumulating evidence from a wide range of studies, including behavioral, cellular, molecular and computational findings, support a key role of dendrites in the encoding and recall of new memories. Dendrites can integrate synaptic inputs in non-linear ways, provide the substrate for local protein synthesis and facilitate the orchestration of signaling pathways that regulate local synaptic plasticity. These capabilities allow them to act as a second layer of computation within the neuron and serve as the fundamental unit of plasticity. As such, dendrites are integral parts of the memory engram, namely the physical representation of memories in the brain and are increasingly studied during learning tasks. Here, we review experimental and computational studies that support a novel, dendritic view of the memory engram that is centered on non-linear dendritic branches as elementary memory units. We highlight the potential implications of dendritic engrams for the learning and memory field and discuss future research directions.
Collapse
Affiliation(s)
- George Kastellakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, Greece
| | - Simone Tasciotti
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, Greece
- Department of Biology, University of Crete, Heraklion, Greece
| | - Ioanna Pandi
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, Greece
- Department of Biology, University of Crete, Heraklion, Greece
| | - Panayiota Poirazi
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion, Greece
| |
Collapse
|
4
|
Rigby M, Grillo FW, Compans B, Neves G, Gallinaro J, Nashashibi S, Horton S, Pereira Machado PM, Carbajal MA, Vizcay-Barrena G, Levet F, Sibarita JB, Kirkland A, Fleck RA, Clopath C, Burrone J. Multi-synaptic boutons are a feature of CA1 hippocampal connections in the stratum oriens. Cell Rep 2023; 42:112397. [PMID: 37074915 PMCID: PMC10695768 DOI: 10.1016/j.celrep.2023.112397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 01/21/2023] [Accepted: 03/30/2023] [Indexed: 04/20/2023] Open
Abstract
Excitatory synapses are typically described as single synaptic boutons (SSBs), where one presynaptic bouton contacts a single postsynaptic spine. Using serial section block-face scanning electron microscopy, we found that this textbook definition of the synapse does not fully apply to the CA1 region of the hippocampus. Roughly half of all excitatory synapses in the stratum oriens involved multi-synaptic boutons (MSBs), where a single presynaptic bouton containing multiple active zones contacted many postsynaptic spines (from 2 to 7) on the basal dendrites of different cells. The fraction of MSBs increased during development (from postnatal day 22 [P22] to P100) and decreased with distance from the soma. Curiously, synaptic properties such as active zone (AZ) or postsynaptic density (PSD) size exhibited less within-MSB variation when compared with neighboring SSBs, features that were confirmed by super-resolution light microscopy. Computer simulations suggest that these properties favor synchronous activity in CA1 networks.
Collapse
Affiliation(s)
- Mark Rigby
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Federico W Grillo
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Benjamin Compans
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Guilherme Neves
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; The Rosalind Franklin Institute, Harwell Campus, Didcot OX11 0FA, UK
| | - Julia Gallinaro
- Bioengineering Department, Imperial College London, London, UK
| | - Sophie Nashashibi
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Sally Horton
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK
| | - Pedro M Pereira Machado
- Centre for Ultrastructural Imaging (CUI), Kings College London, New Hunts House, Guys Hospital Campus, London SE1 1UL, UK
| | - Maria Alejandra Carbajal
- Centre for Ultrastructural Imaging (CUI), Kings College London, New Hunts House, Guys Hospital Campus, London SE1 1UL, UK
| | - Gema Vizcay-Barrena
- Centre for Ultrastructural Imaging (CUI), Kings College London, New Hunts House, Guys Hospital Campus, London SE1 1UL, UK
| | - Florian Levet
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, 33000 Bordeaux, France; University Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UAR3420, US 4, 33000 Bordeaux, France
| | - Jean-Baptiste Sibarita
- University Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, 33000 Bordeaux, France
| | - Angus Kirkland
- The Rosalind Franklin Institute, Harwell Campus, Didcot OX11 0FA, UK
| | - Roland A Fleck
- Centre for Ultrastructural Imaging (CUI), Kings College London, New Hunts House, Guys Hospital Campus, London SE1 1UL, UK
| | - Claudia Clopath
- Bioengineering Department, Imperial College London, London, UK
| | - Juan Burrone
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK; Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, UK.
| |
Collapse
|
5
|
Liu Z, Zhu Y, Zhang L, Jiang W, Liu Y, Tang Q, Cai X, Li J, Wang L, Tao C, Yin X, Li X, Hou S, Jiang D, Liu K, Zhou X, Zhang H, Liu M, Fan C, Tian Y. Structural and functional imaging of brains. Sci China Chem 2022; 66:324-366. [PMID: 36536633 PMCID: PMC9753096 DOI: 10.1007/s11426-022-1408-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/28/2022] [Indexed: 12/23/2022]
Abstract
Analyzing the complex structures and functions of brain is the key issue to understanding the physiological and pathological processes. Although neuronal morphology and local distribution of neurons/blood vessels in the brain have been known, the subcellular structures of cells remain challenging, especially in the live brain. In addition, the complicated brain functions involve numerous functional molecules, but the concentrations, distributions and interactions of these molecules in the brain are still poorly understood. In this review, frontier techniques available for multiscale structure imaging from organelles to the whole brain are first overviewed, including magnetic resonance imaging (MRI), computed tomography (CT), positron emission tomography (PET), serial-section electron microscopy (ssEM), light microscopy (LM) and synchrotron-based X-ray microscopy (XRM). Specially, XRM for three-dimensional (3D) imaging of large-scale brain tissue with high resolution and fast imaging speed is highlighted. Additionally, the development of elegant methods for acquisition of brain functions from electrical/chemical signals in the brain is outlined. In particular, the new electrophysiology technologies for neural recordings at the single-neuron level and in the brain are also summarized. We also focus on the construction of electrochemical probes based on dual-recognition strategy and surface/interface chemistry for determination of chemical species in the brain with high selectivity and long-term stability, as well as electrochemophysiological microarray for simultaneously recording of electrochemical and electrophysiological signals in the brain. Moreover, the recent development of brain MRI probes with high contrast-to-noise ratio (CNR) and sensitivity based on hyperpolarized techniques and multi-nuclear chemistry is introduced. Furthermore, multiple optical probes and instruments, especially the optophysiological Raman probes and fiber Raman photometry, for imaging and biosensing in live brain are emphasized. Finally, a brief perspective on existing challenges and further research development is provided.
Collapse
Affiliation(s)
- Zhichao Liu
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241 China
| | - Ying Zhu
- Interdisciplinary Research Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 201210 China
| | - Liming Zhang
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241 China
| | - Weiping Jiang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Chinese Academy of Sciences, Wuhan National Laboratory for Optoelectronics, Wuhan, 430071 China
| | - Yawei Liu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 China
| | - Qiaowei Tang
- Interdisciplinary Research Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 201210 China
| | - Xiaoqing Cai
- Interdisciplinary Research Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 201210 China
| | - Jiang Li
- Interdisciplinary Research Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 201210 China
| | - Lihua Wang
- Interdisciplinary Research Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 201210 China
| | - Changlu Tao
- Interdisciplinary Center for Brain Information, Brain Cognition and Brain Disease Institute, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055 China
| | | | - Xiaowei Li
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240 China
| | - Shangguo Hou
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, 518055 China
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Kai Liu
- Department of Chemistry, Tsinghua University, Beijing, 100084 China
| | - Xin Zhou
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Chinese Academy of Sciences, Wuhan National Laboratory for Optoelectronics, Wuhan, 430071 China
| | - Hongjie Zhang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 China
- Department of Chemistry, Tsinghua University, Beijing, 100084 China
| | - Maili Liu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Chinese Academy of Sciences, Wuhan National Laboratory for Optoelectronics, Wuhan, 430071 China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, 200240 China
| | - Yang Tian
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241 China
| |
Collapse
|
6
|
Zhang T, Zhang X, Zhu W, Lu Z, Wang Y, Zhang Y. Study on the diversity of mental states and neuroplasticity of the brain during human-machine interaction. Front Neurosci 2022; 16:921058. [PMID: 36570838 PMCID: PMC9768214 DOI: 10.3389/fnins.2022.921058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022] Open
Abstract
Introduction With the increasing demand for human-machine collaboration systems, more and more attention has been paid to the influence of human factors on the performance and security of the entire system. Especially in high-risk, high-precision, and difficult special tasks (such as space station maintenance tasks, anti-terrorist EOD tasks, surgical robot teleoperation tasks, etc.), there are higher requirements for the operator's perception and cognitive level. However, as the human brain is a complex and open giant system, the perception ability and cognitive level of the human are dynamically variable, so that it will seriously affect the performance and security of the whole system. Methods The method proposed in this paper innovatively explained this phenomenon from two dimensions of brain space and time and attributed the dynamic changes of perception, cognitive level, and operational skills to the mental state diversity and the brain neuroplasticity. In terms of the mental state diversity, the mental states evoked paradigm and the functional brain network analysis method during work were proposed. In terms of neuroplasticity, the cognitive training intervention paradigm and the functional brain network analysis method were proposed. Twenty-six subjects participated in the mental state evoked experiment and the cognitive training intervention experiment. Results The results showed that (1) the mental state of the subjects during work had the characteristics of dynamic change, and due to the influence of stimulus conditions and task patterns, the mental state showed diversity. There were significant differences between functional brain networks in different mental states, the information processing efficiency and the mechanism of brain area response had changed significantly. (2) The small-world attributes of the functional brain network of the subjects before and after the cognitive training experiment were significantly different. The brain had adjusted the distribution of information flow and resources, reducing costs and increasing efficiency as a whole. It was demonstrated that the global topology of the cortical connectivity network was reconfigured and neuroplasticity was altered through cognitive training intervention. Discussion In summary, this paper revealed that mental state and neuroplasticity could change the information processing efficiency and the response mechanism of brain area, thus causing the change of perception, cognitive level and operational skills, which provided a theoretical basis for studying the relationship between neural information processing and behavior.
Collapse
Affiliation(s)
- Teng Zhang
- School of Mechanical Engineering, Xi’an Jiaotong University, Xi’an, China
| | - Xiaodong Zhang
- School of Mechanical Engineering, Xi’an Jiaotong University, Xi’an, China
- Shaanxi Key Laboratory of Intelligent Robot, Xi’an Jiaotong University, Xi’an, China
| | - Wenjing Zhu
- School of Mechanical Engineering, Xi’an Jiaotong University, Xi’an, China
| | - Zhufeng Lu
- School of Mechanical Engineering, Xi’an Jiaotong University, Xi’an, China
| | - Yu Wang
- School of Mechanical Engineering, Xi’an Jiaotong University, Xi’an, China
| | - Yingjie Zhang
- School of Mechanical Engineering, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
7
|
Effects of acute exercise on memory: Considerations of exercise intensity, post-exercise recovery period and aerobic endurance. Mem Cognit 2022; 51:1011-1026. [PMID: 36401115 PMCID: PMC9676734 DOI: 10.3758/s13421-022-01373-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2022] [Indexed: 11/19/2022]
Abstract
Accumulating research demonstrates that acute exercise can enhance long-term episodic memory. However, it is unclear if there is an intensity-specific effect of acute exercise on long-term episodic memory function and whether this is influenced by the post-exercise recovery period, which was the primary objective of this experiment. Another uncertainty in the literature is whether aerobic endurance influences the interaction between exercise intensity and post-exercise recovery period on long-term episodic memory function, which was a secondary objective of this study. With exercise intensity and post-exercise recovery period occurring as within-subject factors, and fitness as a between-subject factor, 59 participants (Mage = 20 years) completed 12 primary laboratory visits. These visits included a 20-min bout of exercise (Control, Moderate, and Vigorous), followed by a recovery period (1, 5, 10, and 15 min) and then a word-list episodic memory task, involving an encoding phase and two long-term recall assessments (20-min and 24-h delayed recall). The primary finding from this experiment was that moderate and vigorous-intensity exercise improved memory function when compared to a non-exercise control. A secondary finding was that individuals with higher levels of aerobic endurance, compared to their lesser fit counterparts, had greater memory performance after exercise (moderate or vigorous) when compared to after a control condition. Additionally, individuals with higher levels of aerobic endurance, compared to their lesser fit counterparts, generally performed better on the memory task with longer post-exercise recovery periods. Future research should carefully consider these parameters when evaluating the effects of acute exercise on long-term episodic memory.
Collapse
|
8
|
Barnett SFH, Goult BT. The MeshCODE to scale-visualising synaptic binary information. Front Cell Neurosci 2022; 16:1014629. [PMID: 36467609 PMCID: PMC9716431 DOI: 10.3389/fncel.2022.1014629] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/01/2022] [Indexed: 08/31/2023] Open
Abstract
The Mercator projection map of the world provides a useful, but distorted, view of the relative scale of countries. Current cellular models suffer from a similar distortion. Here, we undertook an in-depth structural analysis of the molecular dimensions in the cell's computational machinery, the MeshCODE, that is assembled from a meshwork of binary switches in the scaffolding proteins talin and vinculin. Talin contains a series of force-dependent binary switches and each domain switching state introduces quantised step-changes in talin length on a micrometre scale. The average dendritic spine is 1 μm in diameter so this analysis identifies a plausible Gearbox-like mechanism for dynamic regulation of synaptic function, whereby the positioning of enzymes and substrates relative to each other, mechanically-encoded by the MeshCODE switch patterns, might control synaptic transmission. Based on biophysical rules and experimentally derived distances, this analysis yields a novel perspective on biological digital information.
Collapse
Affiliation(s)
- Samuel F. H. Barnett
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Benjamin T. Goult
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| |
Collapse
|
9
|
Lillvis JL, Otsuna H, Ding X, Pisarev I, Kawase T, Colonell J, Rokicki K, Goina C, Gao R, Hu A, Wang K, Bogovic J, Milkie DE, Meienberg L, Mensh BD, Boyden ES, Saalfeld S, Tillberg PW, Dickson BJ. Rapid reconstruction of neural circuits using tissue expansion and light sheet microscopy. eLife 2022; 11:e81248. [PMID: 36286237 PMCID: PMC9651950 DOI: 10.7554/elife.81248] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 10/25/2022] [Indexed: 11/13/2022] Open
Abstract
Brain function is mediated by the physiological coordination of a vast, intricately connected network of molecular and cellular components. The physiological properties of neural network components can be quantified with high throughput. The ability to assess many animals per study has been critical in relating physiological properties to behavior. By contrast, the synaptic structure of neural circuits is presently quantifiable only with low throughput. This low throughput hampers efforts to understand how variations in network structure relate to variations in behavior. For neuroanatomical reconstruction, there is a methodological gulf between electron microscopic (EM) methods, which yield dense connectomes at considerable expense and low throughput, and light microscopic (LM) methods, which provide molecular and cell-type specificity at high throughput but without synaptic resolution. To bridge this gulf, we developed a high-throughput analysis pipeline and imaging protocol using tissue expansion and light sheet microscopy (ExLLSM) to rapidly reconstruct selected circuits across many animals with single-synapse resolution and molecular contrast. Using Drosophila to validate this approach, we demonstrate that it yields synaptic counts similar to those obtained by EM, enables synaptic connectivity to be compared across sex and experience, and can be used to correlate structural connectivity, functional connectivity, and behavior. This approach fills a critical methodological gap in studying variability in the structure and function of neural circuits across individuals within and between species.
Collapse
Affiliation(s)
- Joshua L Lillvis
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Hideo Otsuna
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Xiaoyu Ding
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Igor Pisarev
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Takashi Kawase
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Jennifer Colonell
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Konrad Rokicki
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Cristian Goina
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Ruixuan Gao
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
- MIT McGovern Institute for Brain ResearchCambridgeUnited States
- Departments of Chemistry and Biological Sciences, University of Illinois ChicagoChicagoUnited States
| | - Amy Hu
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Kaiyu Wang
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - John Bogovic
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Daniel E Milkie
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | | | - Brett D Mensh
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Edward S Boyden
- MIT McGovern Institute for Brain ResearchCambridgeUnited States
- Howard Hughes Medical InstituteCambridgeUnited States
| | - Stephan Saalfeld
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Paul W Tillberg
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
| | - Barry J Dickson
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnUnited States
- Queensland Brain Institute, The University of QueenslandSt LuciaAustralia
| |
Collapse
|
10
|
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: 1.0] [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
|
11
|
Unique transsynaptic complexes enable long-term synaptic plasticity in a synapse-specific manner. Proc Natl Acad Sci U S A 2022; 119:e2206429119. [PMID: 35737826 PMCID: PMC9271206 DOI: 10.1073/pnas.2206429119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
12
|
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: 2.7] [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
|
13
|
Zhu Y, Uytiepo M, Bushong E, Haberl M, Beutter E, Scheiwe F, Zhang W, Chang L, Luu D, Chui B, Ellisman M, Maximov A. Nanoscale 3D EM reconstructions reveal intrinsic mechanisms of structural diversity of chemical synapses. Cell Rep 2021; 35:108953. [PMID: 33826888 PMCID: PMC8354523 DOI: 10.1016/j.celrep.2021.108953] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/21/2021] [Accepted: 03/15/2021] [Indexed: 10/25/2022] Open
Abstract
Chemical synapses of shared cellular origins have remarkably heterogeneous structures, but how this diversity is generated is unclear. Here, we use three-dimensional (3D) electron microscopy and artificial intelligence algorithms for image processing to reconstruct functional excitatory microcircuits in the mouse hippocampus and microcircuits in which neurotransmitter signaling is permanently suppressed with genetic tools throughout the lifespan. These nanoscale analyses reveal that experience is dispensable for morphogenesis of synapses with different geometric shapes and contents of membrane organelles and that arrangement of morphologically distinct connections in local networks is stochastic. Moreover, loss of activity increases the variability in sizes of opposed pre- and postsynaptic structures without disrupting their alignments, suggesting that inherently variable weights of naive connections become progressively matched with repetitive use. These results demonstrate that mechanisms for the structural diversity of neuronal synapses are intrinsic and provide insights into how circuits essential for memory storage assemble and integrate information.
Collapse
Affiliation(s)
- Yongchuan Zhu
- Department of Neuroscience, The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Marco Uytiepo
- Department of Neuroscience, The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Eric Bushong
- National Center for Microscopy and Imaging Research, University of California, San Diego, CA 92037, USA; Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA 92037, USA
| | - Matthias Haberl
- National Center for Microscopy and Imaging Research, University of California, San Diego, CA 92037, USA; Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA 92037, USA
| | - Elizabeth Beutter
- Department of Neuroscience, The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Frederieke Scheiwe
- Department of Neuroscience, The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Weiheng Zhang
- Department of Neuroscience, The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Lyanne Chang
- Department of Neuroscience, The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Danielle Luu
- Department of Neuroscience, The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Brandon Chui
- Department of Neuroscience, The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Mark Ellisman
- National Center for Microscopy and Imaging Research, University of California, San Diego, CA 92037, USA; Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA 92037, USA.
| | - Anton Maximov
- Department of Neuroscience, The Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
14
|
Reddish FN, Miller CL, Deng X, Dong B, Patel AA, Ghane MA, Mosca B, McBean C, Wu S, Solntsev KM, Zhuo Y, Gadda G, Fang N, Cox DN, Mabb AM, Treves S, Zorzato F, Yang JJ. Rapid subcellular calcium responses and dynamics by calcium sensor G-CatchER . iScience 2021; 24:102129. [PMID: 33665552 PMCID: PMC7900224 DOI: 10.1016/j.isci.2021.102129] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 12/14/2020] [Accepted: 01/26/2021] [Indexed: 12/15/2022] Open
Abstract
The precise spatiotemporal characteristics of subcellular calcium (Ca2+) transients are critical for the physiological processes. Here we report a green Ca2+ sensor called "G-CatchER+" using a protein design to report rapid local ER Ca2+ dynamics with significantly improved folding properties. G-CatchER+ exhibits a superior Ca2+ on rate to G-CEPIA1er and has a Ca2+-induced fluorescence lifetimes increase. G-CatchER+ also reports agonist/antagonist triggered Ca2+ dynamics in several cell types including primary neurons that are orchestrated by IP3Rs, RyRs, and SERCAs with an ability to differentiate expression. Upon localization to the lumen of the RyR channel (G-CatchER+-JP45), we report a rapid local Ca2+ release that is likely due to calsequestrin. Transgenic expression of G-CatchER+ in Drosophila muscle demonstrates its utility as an in vivo reporter of stimulus-evoked SR local Ca2+ dynamics. G-CatchER+ will be an invaluable tool to examine local ER/SR Ca2+ dynamics and facilitate drug development associated with ER dysfunction.
Collapse
Affiliation(s)
- Florence N. Reddish
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| | - Cassandra L. Miller
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| | - Xiaonan Deng
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| | - Bin Dong
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| | - Atit A. Patel
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30303, USA
| | - Mohammad A. Ghane
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30303, USA
| | - Barbara Mosca
- Department of Life Sciences, General Pathology, University of Ferrara, Ferrara, Italy
| | - Cheyenne McBean
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| | - Shengnan Wu
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30303, USA
| | - Kyril M. Solntsev
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - You Zhuo
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| | - Giovanni Gadda
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| | - Ning Fang
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| | - Daniel N. Cox
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30303, USA
| | - Angela M. Mabb
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30303, USA
| | - Susan Treves
- Department of Life Sciences, General Pathology, University of Ferrara, Ferrara, Italy
- Department of Biomedicine, Basel University, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Francesco Zorzato
- Department of Life Sciences, General Pathology, University of Ferrara, Ferrara, Italy
- Department of Biomedicine, Basel University, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Jenny J. Yang
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging Facility, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
15
|
Blazquez-Llorca L, Miguéns M, Montero-Crespo M, Selvas A, Gonzalez-Soriano J, Ambrosio E, DeFelipe J. 3D Synaptic Organization of the Rat CA1 and Alterations Induced by Cocaine Self-Administration. Cereb Cortex 2021; 31:1927-1952. [PMID: 33253368 PMCID: PMC7945021 DOI: 10.1093/cercor/bhaa331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 09/10/2020] [Accepted: 10/12/2020] [Indexed: 12/24/2022] Open
Abstract
The hippocampus plays a key role in contextual conditioning and has been proposed as an important component of the cocaine addiction brain circuit. To gain knowledge about cocaine-induced alterations in this circuit, we used focused ion beam milling/scanning electron microscopy to reveal and quantify the three-dimensional synaptic organization of the neuropil of the stratum radiatum of the rat CA1, under normal circumstances and after cocaine-self administration (SA). Most synapses are asymmetric (excitatory), macular-shaped, and in contact with dendritic spine heads. After cocaine-SA, the size and the complexity of the shape of both asymmetric and symmetric (inhibitory) synapses increased but no changes were observed in the synaptic density. This work constitutes the first detailed report on the 3D synaptic organization in the stratum radiatum of the CA1 field of cocaine-SA rats. Our data contribute to the elucidation of the normal and altered synaptic organization of the hippocampus, which is crucial for better understanding the neurobiological mechanisms underlying cocaine addiction.
Collapse
Affiliation(s)
- L Blazquez-Llorca
- Departamento de Psicobiología, Facultad de Psicología, Universidad Nacional de Educación a Distancia (UNED), 28040 Madrid, Spain.,Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, 28223 Madrid, Spain.,Sección Departamental de Anatomía y Embriología (Veterinaria), Facultad de Veterinaria, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - M Miguéns
- Departamento de Psicología Básica I, Facultad de Psicología, Universidad Nacional de Educación a Distancia (UNED), 28040 Madrid, Spain
| | - M Montero-Crespo
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, 28223 Madrid, Spain.,Instituto Cajal, Consejo Superior de Investigaciones Científicas, 28002 Madrid, Spain
| | - A Selvas
- Departamento de Psicobiología, Facultad de Psicología, Universidad Nacional de Educación a Distancia (UNED), 28040 Madrid, Spain
| | - J Gonzalez-Soriano
- Sección Departamental de Anatomía y Embriología (Veterinaria), Facultad de Veterinaria, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - E Ambrosio
- Departamento de Psicobiología, Facultad de Psicología, Universidad Nacional de Educación a Distancia (UNED), 28040 Madrid, Spain
| | - J DeFelipe
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, 28223 Madrid, Spain.,Instituto Cajal, Consejo Superior de Investigaciones Científicas, 28002 Madrid, Spain
| |
Collapse
|
16
|
Nahirney PC, Tremblay ME. Brain Ultrastructure: Putting the Pieces Together. Front Cell Dev Biol 2021; 9:629503. [PMID: 33681208 PMCID: PMC7930431 DOI: 10.3389/fcell.2021.629503] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 01/20/2021] [Indexed: 12/11/2022] Open
Abstract
Unraveling the fine structure of the brain is important to provide a better understanding of its normal and abnormal functioning. Application of high-resolution electron microscopic techniques gives us an unprecedented opportunity to discern details of the brain parenchyma at nanoscale resolution, although identifying different cell types and their unique features in two-dimensional, or three-dimensional images, remains a challenge even to experts in the field. This article provides insights into how to identify the different cell types in the central nervous system, based on nuclear and cytoplasmic features, amongst other unique characteristics. From the basic distinction between neurons and their supporting cells, the glia, to differences in their subcellular compartments, organelles and their interactions, ultrastructural analyses can provide unique insights into the changes in brain function during aging and disease conditions, such as stroke, neurodegeneration, infection and trauma. Brain parenchyma is composed of a dense mixture of neuronal and glial cell bodies, together with their intertwined processes. Intracellular components that vary between cells, and can become altered with aging or disease, relate to the cytoplasmic and nucleoplasmic density, nuclear heterochromatin pattern, mitochondria, endoplasmic reticulum and Golgi complex, lysosomes, neurosecretory vesicles, and cytoskeletal elements (actin, intermediate filaments, and microtubules). Applying immunolabeling techniques to visualize membrane-bound or intracellular proteins in neurons and glial cells gives an even better appreciation of the subtle differences unique to these cells across contexts of health and disease. Together, our observations reveal how simple ultrastructural features can be used to identify specific changes in cell types, their health status, and functional relationships in the brain.
Collapse
|
17
|
Liu Q, Sun YM, Huang H, Chen C, Wan J, Ma LH, Sun YY, Miao HH, Wu YQ. Sirtuin 3 protects against anesthesia/surgery-induced cognitive decline in aged mice by suppressing hippocampal neuroinflammation. J Neuroinflammation 2021; 18:41. [PMID: 33541361 PMCID: PMC7863360 DOI: 10.1186/s12974-021-02089-z] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 01/19/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Postoperative cognitive dysfunction (POCD) is a very common complication that might increase the morbidity and mortality of elderly patients after surgery. However, the mechanism of POCD remains largely unknown. The NAD-dependent deacetylase protein Sirtuin 3 (SIRT3) is located in the mitochondria and regulates mitochondrial function. SIRT3 is the only sirtuin that specifically plays a role in extending lifespan in humans and is associated with neurodegenerative diseases. Therefore, the aim of this study was to evaluate the effect of SIRT3 on anesthesia/surgery-induced cognitive impairment in aged mice. METHODS SIRT3 expression levels were decreased after surgery. For the interventional study, an adeno-associated virus (AAV)-SIRT3 vector or an empty vector was microinjected into hippocampal CA1 region before anesthesia/surgery. Western blotting, immunofluorescence staining, and enzyme-linked immune-sorbent assay (ELISA) were used to measure the oxidative stress response and downstream microglial activation and proinflammatory cytokines, and Golgi staining and long-term potentiation (LTP) recording were applied to evaluate synaptic plasticity. RESULTS Overexpression of SIRT3 in the CA1 region attenuated anesthesia/surgery-induced learning and memory dysfunction as well as synaptic plasticity dysfunction and the oxidative stress response (superoxide dismutase [SOD] and malondialdehyde [MDA]) in aged mice with POCD. In addition, microglia activation (ionized calcium binding adapter molecule 1 [Iba1]) and neuroinflammatory cytokine levels (tumor necrosis factor-alpha [TNF-α], interleukin [IL]-1β and IL-6) were regulated after anesthesia/surgery in a SIRT3-dependent manner. CONCLUSION The results of the current study demonstrate that SIRT3 has a critical effect in the mechanism of POCD in aged mice by suppressing hippocampal neuroinflammation and reveal that SIRT3 may be a promising therapeutic and diagnostic target for POCD.
Collapse
Affiliation(s)
- Qiang Liu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Tongshan Road 209, Xuzhou, 221004, P.R. China
| | - Yi-Man Sun
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Tongshan Road 209, Xuzhou, 221004, P.R. China
| | - Hui Huang
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Tongshan Road 209, Xuzhou, 221004, P.R. China
| | - Chen Chen
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Tongshan Road 209, Xuzhou, 221004, P.R. China
| | - Jie Wan
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Tongshan Road 209, Xuzhou, 221004, P.R. China
| | - Lin-Hui Ma
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Tongshan Road 209, Xuzhou, 221004, P.R. China
| | - Yin-Ying Sun
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Tongshan Road 209, Xuzhou, 221004, P.R. China
| | - Hui-Hui Miao
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, P.R. China.
| | - Yu-Qing Wu
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Tongshan Road 209, Xuzhou, 221004, P.R. China.
| |
Collapse
|
18
|
Colombo MN, Maiellano G, Putignano S, Scandella L, Francolini M. Comparative 2D and 3D Ultrastructural Analyses of Dendritic Spines from CA1 Pyramidal Neurons in the Mouse Hippocampus. Int J Mol Sci 2021; 22:ijms22031188. [PMID: 33530380 PMCID: PMC7865959 DOI: 10.3390/ijms22031188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/19/2021] [Accepted: 01/22/2021] [Indexed: 11/21/2022] Open
Abstract
Three-dimensional (3D) reconstruction from electron microscopy (EM) datasets is a widely used tool that has improved our knowledge of synapse ultrastructure and organization in the brain. Rearrangements of synapse structure following maturation and in synaptic plasticity have been broadly described and, in many cases, the defective architecture of the synapse has been associated to functional impairments. It is therefore important, when studying brain connectivity, to map these rearrangements with the highest accuracy possible, considering the affordability of the different EM approaches to provide solid and reliable data about the structure of such a small complex. The aim of this work is to compare quantitative data from two dimensional (2D) and 3D EM of mouse hippocampal CA1 (apical dendrites), to define whether the results from the two approaches are consistent. We examined asymmetric excitatory synapses focusing on post synaptic density and dendritic spine area and volume as well as spine density, and we compared the results obtained with the two methods. The consistency between the 2D and 3D results questions the need—for many applications—of using volumetric datasets (costly and time consuming in terms of both acquisition and analysis), with respect to the more accessible measurements from 2D EM projections.
Collapse
|
19
|
Abstract
Unraveling the fine structure of the brain is important to provide a better understanding of its normal and abnormal functioning. Application of high-resolution electron microscopic techniques gives us an unprecedented opportunity to discern details of the brain parenchyma at nanoscale resolution, although identifying different cell types and their unique features in two-dimensional, or three-dimensional images, remains a challenge even to experts in the field. This article provides insights into how to identify the different cell types in the central nervous system, based on nuclear and cytoplasmic features, amongst other unique characteristics. From the basic distinction between neurons and their supporting cells, the glia, to differences in their subcellular compartments, organelles and their interactions, ultrastructural analyses can provide unique insights into the changes in brain function during aging and disease conditions, such as stroke, neurodegeneration, infection and trauma. Brain parenchyma is composed of a dense mixture of neuronal and glial cell bodies, together with their intertwined processes. Intracellular components that vary between cells, and can become altered with aging or disease, relate to the cytoplasmic and nucleoplasmic density, nuclear heterochromatin pattern, mitochondria, endoplasmic reticulum and Golgi complex, lysosomes, neurosecretory vesicles, and cytoskeletal elements (actin, intermediate filaments, and microtubules). Applying immunolabeling techniques to visualize membrane-bound or intracellular proteins in neurons and glial cells gives an even better appreciation of the subtle differences unique to these cells across contexts of health and disease. Together, our observations reveal how simple ultrastructural features can be used to identify specific changes in cell types, their health status, and functional relationships in the brain.
Collapse
Affiliation(s)
- Patrick C Nahirney
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Marie-Eve Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| |
Collapse
|
20
|
Montero-Crespo M, Dominguez-Alvaro M, Rondon-Carrillo P, Alonso-Nanclares L, DeFelipe J, Blazquez-Llorca L. Three-dimensional synaptic organization of the human hippocampal CA1 field. eLife 2020; 9:e57013. [PMID: 32690133 PMCID: PMC7375818 DOI: 10.7554/elife.57013] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022] Open
Abstract
The hippocampal CA1 field integrates a wide variety of subcortical and cortical inputs, but its synaptic organization in humans is still unknown due to the difficulties involved studying the human brain via electron microscope techniques. However, we have shown that the 3D reconstruction method using Focused Ion Beam/Scanning Electron Microscopy (FIB/SEM) can be applied to study in detail the synaptic organization of the human brain obtained from autopsies, yielding excellent results. Using this technology, 24,752 synapses were fully reconstructed in CA1, revealing that most of them were excitatory, targeting dendritic spines and displaying a macular shape, regardless of the layer examined. However, remarkable differences were observed between layers. These data constitute the first extensive description of the synaptic organization of the neuropil of the human CA1 region.
Collapse
Affiliation(s)
- Marta Montero-Crespo
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC)MadridSpain
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de MadridMadridSpain
| | - Marta Dominguez-Alvaro
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de MadridMadridSpain
| | - Patricia Rondon-Carrillo
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de MadridMadridSpain
| | - Lidia Alonso-Nanclares
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC)MadridSpain
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de MadridMadridSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIIIMadridSpain
| | - Javier DeFelipe
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC)MadridSpain
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de MadridMadridSpain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIIIMadridSpain
| | - Lidia Blazquez-Llorca
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de MadridMadridSpain
- Departamento de Psicobiología, Facultad de Psicología, Universidad Nacional de Educación a Distancia (UNED)MadridSpain
| |
Collapse
|
21
|
Wang S, Yao H, Xu Y, Hao R, Zhang W, Liu H, Huang Y, Guo W, Lu B. Therapeutic potential of a TrkB agonistic antibody for Alzheimer's disease. Theranostics 2020; 10:6854-6874. [PMID: 32550908 PMCID: PMC7295064 DOI: 10.7150/thno.44165] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/11/2020] [Indexed: 12/26/2022] Open
Abstract
Repeated failures of "Aβ-lowering" therapies call for new targets and therapeutic approaches for Alzheimer's disease (AD). We propose to treat AD by halting neuronal death and repairing synapses using a BDNF-based therapy. To overcome the poor druggability of BDNF, we have developed an agonistic antibody AS86 to mimic the function of BDNF, and evaluate its therapeutic potential for AD. Method: Biochemical, electrophysiological and behavioral techniques were used to investigate the effects of AS86 in vitro and in vivo. Results: AS86 specifically activated the BDNF receptor TrkB and its downstream signaling, without affecting its other receptor p75NTR. It promoted neurite outgrowth, enhanced spine growth and prevented Aβ-induced cell death in cultured neurons, and facilitated Long-Term Potentiation (LTP) in hippocampal slices. A single-dose tail-vein injection of AS86 activated TrkB signaling in the brain, with a half-life of 6 days in the blood and brain. Bi-weekly peripheral administration of AS86 rescued the deficits in object-recognition memory in the APP/PS1 mouse model. AS86 also reversed spatial memory deficits in the 11-month, but not 14-month old AD mouse model. Conclusion: These results demonstrate the potential of AS86 in AD therapy, suggesting that neuronal and/or synaptic repair as an alternative therapeutic strategy for AD.
Collapse
Affiliation(s)
- Shudan Wang
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China, 100084
- Beijing Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China, 100070
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China, 518057
| | - Hongyang Yao
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China, 100084
| | - Yihua Xu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China, 100084
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China, 518057
| | - Rui Hao
- Center of Translational Medicine, Tongji Hospital, Tongji University School of Medicine, Shanghai, China, 200065
| | - Wen Zhang
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China, 100084
| | - Hang Liu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China, 100084
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China, 518057
| | - Ying Huang
- Center of Translational Medicine, Tongji Hospital, Tongji University School of Medicine, Shanghai, China, 200065
| | - Wei Guo
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China, 100084
- Beijing Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China, 100070
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China, 518057
| | - Bai Lu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China, 100084
- Beijing Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China, 100070
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China, 518057
| |
Collapse
|
22
|
Ultrastructure of light-activated axons following optogenetic stimulation to produce late-phase long-term potentiation. PLoS One 2020; 15:e0226797. [PMID: 31940316 PMCID: PMC6961864 DOI: 10.1371/journal.pone.0226797] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 12/04/2019] [Indexed: 12/03/2022] Open
Abstract
Analysis of neuronal compartments has revealed many state-dependent changes in geometry but establishing synapse-specific mechanisms at the nanoscale has proven elusive. We co-expressed channelrhodopsin2-GFP and mAPEX2 in a subset of hippocampal CA3 neurons and used trains of light to induce late-phase long-term potentiation (L-LTP) in area CA1. L-LTP was shown to be specific to the labeled axons by severing CA3 inputs, which prevented back-propagating recruitment of unlabeled axons. Membrane-associated mAPEX2 tolerated microwave-enhanced chemical fixation and drove tyramide signal amplification to deposit Alexa Fluor dyes in the light-activated axons. Subsequent post-embedding immunogold labeling resulted in outstanding ultrastructure and clear distinctions between labeled (activated), and unlabeled axons without obscuring subcellular organelles. The gold-labeled axons in potentiated slices were reconstructed through serial section electron microscopy; presynaptic vesicles and other constituents could be quantified unambiguously. The genetic specification, reliable physiology, and compatibility with established methods for ultrastructural preservation make this an ideal approach to link synapse ultrastructure and function in intact circuits.
Collapse
|
23
|
Kulik YD, Watson DJ, Cao G, Kuwajima M, Harris KM. Structural plasticity of dendritic secretory compartments during LTP-induced synaptogenesis. eLife 2019; 8:e46356. [PMID: 31433297 PMCID: PMC6728136 DOI: 10.7554/elife.46356] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 08/20/2019] [Indexed: 12/30/2022] Open
Abstract
Long-term potentiation (LTP), an increase in synaptic efficacy following high-frequency stimulation, is widely considered a mechanism of learning. LTP involves local remodeling of dendritic spines and synapses. Smooth endoplasmic reticulum (SER) and endosomal compartments could provide local stores of membrane and proteins, bypassing the distant Golgi apparatus. To test this hypothesis, effects of LTP were compared to control stimulation in rat hippocampal area CA1 at postnatal day 15 (P15). By two hours, small spines lacking SER increased after LTP, whereas large spines did not change in frequency, size, or SER content. Total SER volume decreased after LTP consistent with transfer of membrane to the added spines. Shaft SER remained more abundant in spiny than aspiny dendritic regions, apparently supporting the added spines. Recycling endosomes were elevated specifically in small spines after LTP. These findings suggest local secretory trafficking contributes to LTP-induced synaptogenesis and primes the new spines for future plasticity.
Collapse
Affiliation(s)
- Yelena D Kulik
- Center for Learning and Memory, Department of NeuroscienceThe University of Texas at AustinAustinUnited States
| | - Deborah J Watson
- Center for Learning and Memory, Department of NeuroscienceThe University of Texas at AustinAustinUnited States
| | - Guan Cao
- Center for Learning and Memory, Department of NeuroscienceThe University of Texas at AustinAustinUnited States
| | - Masaaki Kuwajima
- Center for Learning and Memory, Department of NeuroscienceThe University of Texas at AustinAustinUnited States
| | - Kristen M Harris
- Center for Learning and Memory, Department of NeuroscienceThe University of Texas at AustinAustinUnited States
| |
Collapse
|
24
|
Balancing Extrasynaptic Excitation and Synaptic Inhibition within Olfactory Bulb Glomeruli. eNeuro 2019; 6:ENEURO.0247-19.2019. [PMID: 31345999 PMCID: PMC6709216 DOI: 10.1523/eneuro.0247-19.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/15/2019] [Accepted: 07/22/2019] [Indexed: 12/31/2022] Open
Abstract
Glutamatergic transmission in the brain typically occurs at well-defined synaptic connections, but increasing evidence indicates that neural excitation can also occur through activation of “extrasynaptic” glutamate receptors. Here, we investigated the underlying mechanisms and functional properties of extrasynaptic signals that are part of a feedforward path of information flow in the olfactory bulb. This pathway involves glutamatergic interneurons, external tufted cells (eTCs), that are excited by olfactory sensory neurons (OSNs) and in turn excite output mitral cells (MCs) extrasynaptically. Using pair-cell and triple-cell recordings in rat bulb slices (of either sex), combined with ultrastructural approaches, we first present evidence that eTC-to-MC signaling results from “spillover” of glutamate released at eTC synapses onto GABAergic periglomerular (PG) cells in glomeruli. Thus, feedforward excitation is an indirect result of and must cooccur with activation of inhibitory circuitry. Next, to examine the dynamics of the competing signals, we assayed the relationship between the number of spikes in eTCs and excitation of MCs or PG cells in pair-cell recordings. This showed that extrasynaptic excitation in MCs is very weak due to single spikes but rises sharply and supralinearly with increasing spikes, differing from sublinear behavior for synaptic excitation of PG cells. Similar dynamics leading to a preference for extrasynaptic excitation were also observed during recordings of extrasynaptic and inhibitory currents in response to OSN input of increasing magnitude. The observed alterations in the balance between extrasynaptic excitation and inhibition in glomeruli with stimulus strength could underlie an intraglomerular mechanism for olfactory contrast enhancement.
Collapse
|
25
|
Class IIa HDACs regulate learning and memory through dynamic experience-dependent repression of transcription. Nat Commun 2019; 10:3469. [PMID: 31375688 PMCID: PMC6677776 DOI: 10.1038/s41467-019-11409-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 07/13/2019] [Indexed: 01/08/2023] Open
Abstract
The formation of new memories requires transcription. However, the mechanisms that limit signaling of relevant gene programs in space and time for precision of information coding remain poorly understood. We found that, during learning, the cellular patterns of expression of early response genes (ERGs) are regulated by class IIa HDACs 4 and 5, transcriptional repressors that transiently enter neuronal nuclei from cytoplasm after sensory input. Mice lacking these repressors in the forebrain have abnormally broad experience-dependent expression of ERGs, altered synaptic architecture and function, elevated anxiety, and severely impaired memory. By acutely manipulating the nuclear activity of class IIa HDACs in behaving animals using a chemical-genetic technique, we further demonstrate that rapid induction of transcriptional programs is critical for memory acquisition but these programs may become dispensable when a stable memory is formed. These results provide new insights into the molecular basis of memory storage. The molecular mechanisms of memory storage remain poorly understood. In this study, authors describe a new mechanism that regulates the cellular patterns of early response gene signaling during learning via the recruitment of two functionally redundant nuclear repressors, class IIa histone deacetylases (HDACs) 4 and 5
Collapse
|
26
|
Chidambaram SB, Rathipriya AG, Bolla SR, Bhat A, Ray B, Mahalakshmi AM, Manivasagam T, Thenmozhi AJ, Essa MM, Guillemin GJ, Chandra R, Sakharkar MK. Dendritic spines: Revisiting the physiological role. Prog Neuropsychopharmacol Biol Psychiatry 2019; 92:161-193. [PMID: 30654089 DOI: 10.1016/j.pnpbp.2019.01.005] [Citation(s) in RCA: 156] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 01/04/2019] [Accepted: 01/12/2019] [Indexed: 12/11/2022]
Abstract
Dendritic spines are small, thin, specialized protrusions from neuronal dendrites, primarily localized in the excitatory synapses. Sophisticated imaging techniques revealed that dendritic spines are complex structures consisting of a dense network of cytoskeletal, transmembrane and scaffolding molecules, and numerous surface receptors. Molecular signaling pathways, mainly Rho and Ras family small GTPases pathways that converge on actin cytoskeleton, regulate the spine morphology and dynamics bi-directionally during synaptic activity. During synaptic plasticity the number and shapes of dendritic spines undergo radical reorganizations. Long-term potentiation (LTP) induction promote spine head enlargement and the formation and stabilization of new spines. Long-term depression (LTD) results in their shrinkage and retraction. Reports indicate increased spine density in the pyramidal neurons of autism and Fragile X syndrome patients and reduced density in the temporal gyrus loci of schizophrenic patients. Post-mortem reports of Alzheimer's brains showed reduced spine number in the hippocampus and cortex. This review highlights the spine morphogenesis process, the activity-dependent structural plasticity and mechanisms by which synaptic activity sculpts the dendritic spines, the structural and functional changes in spines during learning and memory using LTP and LTD processes. It also discusses on spine status in neurodegenerative diseases and the impact of nootropics and neuroprotective agents on the functional restoration of dendritic spines.
Collapse
Affiliation(s)
- Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSSAHER), Mysuru, Karnataka 570015, India.
| | - A G Rathipriya
- Food and Brain Research Foundation, Chennai, Tamil Nadu, India
| | - Srinivasa Rao Bolla
- Department of Anatomy, College of Medicine, Imam Abdulrahman Bin Faisal University, Damam, Saudi Arabia
| | - Abid Bhat
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSSAHER), Mysuru, Karnataka 570015, India
| | - Bipul Ray
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSSAHER), Mysuru, Karnataka 570015, India
| | - Arehally Marappa Mahalakshmi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSSAHER), Mysuru, Karnataka 570015, India
| | - Thamilarasan Manivasagam
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamilnadu, India
| | - Arokiasamy Justin Thenmozhi
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Annamalainagar, Tamilnadu, India
| | - Musthafa Mohamed Essa
- Department of Food Science and Nutrition, CAMS, Sultan Qaboos University, Muscat, Oman
| | - Gilles J Guillemin
- Neuropharmacology Group, Faculty of Medicine and Health Sciences, Deb Bailey MND Research Laboratory, Macquarie University, Sydney, NSW 2109, Australia
| | - Ramesh Chandra
- Department of Chemistry, Ambedkar Centre for BioMedical Research, Delhi University, Delhi 110007, India
| | - Meena Kishore Sakharkar
- College of Pharmacy and Nutrition, University of Saskatchewan, 107, Wiggins Road, Saskatoon, SK S7N 5C9, Canada.
| |
Collapse
|
27
|
Metzbower SR, Joo Y, Benavides DR, Blanpied TA. Properties of Individual Hippocampal Synapses Influencing NMDA-Receptor Activation by Spontaneous Neurotransmission. eNeuro 2019; 6:ENEURO.0419-18.2019. [PMID: 31110134 PMCID: PMC6541874 DOI: 10.1523/eneuro.0419-18.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 05/05/2019] [Accepted: 05/12/2019] [Indexed: 12/14/2022] Open
Abstract
NMDA receptor (NMDAR) activation is critical for maintenance and modification of synapse strength. Specifically, NMDAR activation by spontaneous glutamate release has been shown to mediate some forms of synaptic plasticity as well as synaptic development. Interestingly, there is evidence that within individual synapses each release mode may be segregated such that postsynaptically there are distinct pools of responsive receptors. To examine potential regulators of NMDAR activation because of spontaneous glutamate release in cultured hippocampal neurons, we used GCaMP6f imaging at single synapses in concert with confocal and super-resolution imaging. Using these single-spine approaches, we found that Ca2+ entry activated by spontaneous release tends to be carried by GluN2B-NMDARs. Additionally, the amount of NMDAR activation varies greatly both between synapses and within synapses, and is unrelated to spine and synapse size, but does correlate loosely with synapse distance from the soma. Despite the critical role of spontaneous activation of NMDARs in maintaining synaptic function, their activation seems to be controlled factors other than synapse size or synapse distance from the soma. It is most likely that NMDAR activation by spontaneous release influenced variability in subsynaptic receptor position, release site position, vesicle content, and channel properties. Therefore, spontaneous activation of NMDARs appears to be regulated distinctly from other receptor types, notably AMPARs, within individual synapses.
Collapse
Affiliation(s)
| | - Yuyoung Joo
- Department of Neurology, University of Maryland School of Medicine, Baltimore, Maryland, 21201
| | - David R Benavides
- Department of Neurology, University of Maryland School of Medicine, Baltimore, Maryland, 21201
| | | |
Collapse
|
28
|
Valtcheva S, Venance L. Control of Long-Term Plasticity by Glutamate Transporters. Front Synaptic Neurosci 2019; 11:10. [PMID: 31024287 PMCID: PMC6465798 DOI: 10.3389/fnsyn.2019.00010] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 03/12/2019] [Indexed: 12/11/2022] Open
Abstract
Activity-dependent long-term changes in synaptic strength constitute key elements for learning and memory formation. Long-term plasticity can be induced in vivo and ex vivo by various physiologically relevant activity patterns. Depending on their temporal statistics, such patterns can induce long-lasting changes in the synaptic weight by potentiating or depressing synaptic transmission. At excitatory synapses, glutamate uptake operated by excitatory amino acid transporters (EAATs) has a critical role in regulating the strength and the extent of receptor activation by afferent activity. EAATs tightly control synaptic transmission and glutamate spillover. EAATs activity can, therefore, determine the polarity and magnitude of long-term plasticity by regulating the spatiotemporal profile of the glutamate transients and thus, the glutamate access to pre- and postsynaptic receptors. Here, we summarize compelling evidence that EAATs regulate various forms of long-term synaptic plasticity and the consequences of such regulation for behavioral output. We speculate that experience-dependent plasticity of EAATs levels can determine the sensitivity of synapses to frequency- or time-dependent plasticity paradigms. We propose that EAATs contribute to the gating of relevant inputs eligible to induce long-term plasticity and thereby select the operating learning rules that match the physiological function of the synapse adapted to the behavioral context.
Collapse
Affiliation(s)
- Silvana Valtcheva
- Dynamics and Pathophysiology of Neuronal Networks Team, Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR7241/INSERM U1050, Paris, France
| | - Laurent Venance
- Dynamics and Pathophysiology of Neuronal Networks Team, Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR7241/INSERM U1050, Paris, France
| |
Collapse
|
29
|
Bagheri A, Habibzadeh P, Razavipour SF, Volmar CH, Chee NT, Brothers SP, Wahlestedt C, Mowla SJ, Faghihi MA. HDAC Inhibitors Induce BDNF Expression and Promote Neurite Outgrowth in Human Neural Progenitor Cells-Derived Neurons. Int J Mol Sci 2019; 20:E1109. [PMID: 30841499 PMCID: PMC6429164 DOI: 10.3390/ijms20051109] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 02/19/2019] [Accepted: 02/28/2019] [Indexed: 12/11/2022] Open
Abstract
Besides its key role in neural development, brain-derived neurotrophic factor (BDNF) is important for long-term potentiation and neurogenesis, which makes it a critical factor in learning and memory. Due to the important role of BDNF in synaptic function and plasticity, an in-house epigenetic library was screened against human neural progenitor cells (HNPCs) and WS1 human skin fibroblast cells using Cell-to-Ct assay kit to identify the small compounds capable of modulating the BDNF expression. In addition to two well-known hydroxamic acid-based histone deacetylase inhibitors (hb-HDACis), SAHA and TSA, several structurally similar HDAC inhibitors including SB-939, PCI-24781 and JNJ-26481585 with even higher impact on BDNF expression, were discovered in this study. Furthermore, by using well-developed immunohistochemistry assays, the selected compounds were also proved to have neurogenic potential improving the neurite outgrowth in HNPCs-derived neurons. In conclusion, we proved the neurogenic potential of several hb-HDACis, alongside their ability to enhance BDNF expression, which by modulating the neurogenesis and/or compensating for neuronal loss, could be propitious for treatment of neurological disorders.
Collapse
Affiliation(s)
- Amir Bagheri
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, P.O. Box 14115-111, Iran.
- Center for Therapeutic Innovation and Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Parham Habibzadeh
- Persian BayanGene Research and Training Center, Shiraz, P.O. Box 7134767617, Iran.
| | - Seyedeh Fatemeh Razavipour
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Claude-Henry Volmar
- Center for Therapeutic Innovation and Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Nancy T Chee
- Center for Therapeutic Innovation and Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Shaun P Brothers
- Center for Therapeutic Innovation and Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Claes Wahlestedt
- Center for Therapeutic Innovation and Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Seyed Javad Mowla
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, P.O. Box 14115-111, Iran.
| | - Mohammad Ali Faghihi
- Center for Therapeutic Innovation and Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Persian BayanGene Research and Training Center, Shiraz, P.O. Box 7134767617, Iran.
| |
Collapse
|
30
|
Borczyk M, Śliwińska MA, Caly A, Bernas T, Radwanska K. Neuronal plasticity affects correlation between the size of dendritic spine and its postsynaptic density. Sci Rep 2019; 9:1693. [PMID: 30737431 PMCID: PMC6368589 DOI: 10.1038/s41598-018-38412-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 11/19/2018] [Indexed: 01/23/2023] Open
Abstract
Structural plasticity of dendritic spines is thought to underlie memory formation. Size of a dendritic spine is considered proportional to the size of its postsynaptic density (PSD), number of glutamate receptors and synaptic strength. However, whether this correlation is true for all dendritic spine volumes, and remains stable during synaptic plasticity, is largely unknown. In this study, we take advantage of 3D electron microscopy and reconstruct dendritic spines and cores of PSDs from the stratum radiatum of the area CA1 of organotypic hippocampal slices. We observe that approximately 1/3 of dendritic spines, in a range of medium sizes, fail to reach significant correlation between dendritic spine volume and PSD surface area or PSD-core volume. During NMDA receptor-dependent chemical long-term potentiation (NMDAR-cLTP) dendritic spines and their PSD not only grow, but also PSD area and PSD-core volume to spine volume ratio is increased, and the correlation between the sizes of these two is tightened. Further analysis specified that only spines that contain smooth endoplasmic reticulum (SER) grow during cLTP, while PSD-cores grow irrespectively of the presence of SER in the spine. Dendritic spines with SER also show higher correlation of the volumetric parameters than spines without SER, and this correlation is further increased during cLTP only in the spines that contain SER. Overall, we found that correlation between PSD surface area and spine volume is not consistent across all spine volumes, is modified and tightened during synaptic plasticity and regulated by SER.
Collapse
Affiliation(s)
- Malgorzata Borczyk
- Laboratory of Molecular Basis of Behavior, the Nencki Institute of Experimental Biology of Polish Academy of Sciences, ul. L. Pasteura 3, Warsaw, 02-093, Poland
| | - Małgorzata Alicja Śliwińska
- Laboratory of Molecular Basis of Behavior, the Nencki Institute of Experimental Biology of Polish Academy of Sciences, ul. L. Pasteura 3, Warsaw, 02-093, Poland.,Laboratory of Imaging Tissue Structure and Function, the Nencki Institute of Experimental Biology of Polish Academy of Sciences, ul. L. Pasteura 3, Warsaw, 02-093, Poland
| | - Anna Caly
- Laboratory of Molecular Basis of Behavior, the Nencki Institute of Experimental Biology of Polish Academy of Sciences, ul. L. Pasteura 3, Warsaw, 02-093, Poland
| | - Tytus Bernas
- Laboratory of Imaging Tissue Structure and Function, the Nencki Institute of Experimental Biology of Polish Academy of Sciences, ul. L. Pasteura 3, Warsaw, 02-093, Poland
| | - Kasia Radwanska
- Laboratory of Molecular Basis of Behavior, the Nencki Institute of Experimental Biology of Polish Academy of Sciences, ul. L. Pasteura 3, Warsaw, 02-093, Poland.
| |
Collapse
|
31
|
Synaptic Plasticity of Human Umbilical Cord Mesenchymal Stem Cell Differentiating into Neuron-like Cells In Vitro Induced by Edaravone. Stem Cells Int 2018; 2018:5304279. [PMID: 30510585 PMCID: PMC6230402 DOI: 10.1155/2018/5304279] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/02/2018] [Accepted: 08/14/2018] [Indexed: 12/12/2022] Open
Abstract
Objective The human umbilical cord mesenchymal stem cells (hUMSCs) are characterized with the potential ability to differentiate to several types of cells. Edaravone has been demonstrated to prevent the hUMSCs from the oxidative damage, especially its ability in antioxidative stress. We hypothesized that Edaravone induces the hUMSCs into the neuron-like cells. Methods The hUMSCs were obtained from the human umbilical cord tissue. The differentiation of hUMSCs was induced by Edaravone with three different doses: 0.65 mg/ml, 1.31 mg/ml, and 2.62 mg/ml. Flow cytometry was used to detect the cell markers. Protein and mRNA levels of nestin, neuron-specific enolase (NSE), and glial fibrillary acidic protein (GFAP) were detected by Western blot and RT-PCR. The expression of synaptophysin (SYN), growth-associated protein 43 (GAP43), and postsynaptic density 95 (PSD95) was detected by Real-Time PCR. Results As long as the prolongation of the culture, the hUMSCs displayed with the long strips or long fusiform to fat and then characterized with the radial helix growth. By using flow cytometry, the cultured hUMSCs at the 3rd, 5th, and 10th passages were expressed with CD73, CD90, and CD105 but not CD11b, CD19, CD34, CD45, and HLA-DR. Most of the hUMSCs cultured with Edaravone exhibited typical nerve-immediately characters including the cell body contraction, increased refraction, and protruding one or more elongated protrusions, which were not found in the control group without addition of Edaravone. NSE, nestin, and GFAP were positive in these neuron-like cells. Edaravone dose-dependently increased expression levels of NSE, nestin, and GFAP. After replacement of maintenance fluid, neuron-like cells continued to be cultured for five days. These neuron-like cells were positive for SYN, PSD95, and GAP43. Conclusion Edaravone can dose-dependently induce hUMSCs to differentiate into neuron-like cells that expressed the neuronal markers including NSE, nestin, and GFAP and synaptic makers such as SYN, PSD95, and GAP43.
Collapse
|
32
|
Urban BE, Xiao L, Dong B, Chen S, Kozorovitskiy Y, Zhang HF. Imaging neuronal structure dynamics using 2-photon super-resolution patterned excitation reconstruction microscopy. JOURNAL OF BIOPHOTONICS 2018; 11:10.1002/jbio.201700171. [PMID: 28976633 PMCID: PMC7313398 DOI: 10.1002/jbio.201700171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 08/31/2017] [Accepted: 09/28/2017] [Indexed: 05/11/2023]
Abstract
Visualizing fine neuronal structures deep inside strongly light-scattering brain tissue remains a challenge in neuroscience. Recent nanoscopy techniques have reached the necessary resolution but often suffer from limited imaging depth, long imaging time or high light fluence requirements. Here, we present two-photon super-resolution patterned excitation reconstruction (2P-SuPER) microscopy for 3-dimensional imaging of dendritic spine dynamics at a maximum demonstrated imaging depth of 130 μm in living brain tissue with approximately 100 nm spatial resolution. We confirmed 2P-SuPER resolution using fluorescence nanoparticle and quantum dot phantoms and imaged spiny neurons in acute brain slices. We induced hippocampal plasticity and showed that 2P-SuPER can resolve increases in dendritic spine head sizes on CA1 pyramidal neurons following theta-burst stimulation of Schaffer collateral axons. 2P-SuPER further revealed nanoscopic increases in dendritic spine neck widths, a feature of synaptic plasticity that has not been thoroughly investigated due to the combined limit of resolution and penetration depth in existing imaging technologies.
Collapse
Affiliation(s)
- Ben E. Urban
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Lei Xiao
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Biqin Dong
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Siyu Chen
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | | | - Hao F. Zhang
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
33
|
Loprinzi PD, Sng E, Frith E. 'Memorcise': implications for patient compliance and medication adherence. PHYSICIAN SPORTSMED 2018; 46:21-23. [PMID: 29111867 DOI: 10.1080/00913847.2018.1402664] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Paul D Loprinzi
- a Department of Health, Exercise Science, and Recreation Management , The University of Mississippi , Oxford , MS , USA
| | - Eveleen Sng
- a Department of Health, Exercise Science, and Recreation Management , The University of Mississippi , Oxford , MS , USA
| | - Emily Frith
- a Department of Health, Exercise Science, and Recreation Management , The University of Mississippi , Oxford , MS , USA
| |
Collapse
|
34
|
Lømo T. Discovering long-term potentiation (LTP) - recollections and reflections on what came after. Acta Physiol (Oxf) 2018; 222. [PMID: 28719040 DOI: 10.1111/apha.12921] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/17/2017] [Accepted: 07/11/2017] [Indexed: 01/31/2023]
Abstract
Chance events led me to a lifelong career in scientific research. They paved the way for being the first to see long-term potentiation of synaptic efficiency (LTP) in Per Andersen's laboratory in Oslo in 1966. Here I describe my way to this discovery and the experiments with Tim Bliss in 1968-1969 that led to Bliss and Lømo, 1973. Surprisingly, we later failed to reproduce these results. I discuss possible reasons for this failure, which made us both leave LTP research, in my case for good, in Tim's case for several years. After 30 years of work in a different field, I renewed my interest in the hippocampus and LTP in the early 2000s and published, for the first time, results that I had obtained 40 years earlier. Here I present my take on how interest in and research on LTP evolved after the early years. This includes a discussion of the functions of hippocampus as seen in those early days, the case of patient H.M., Donald Hebb's place in the story, the search for 'memory molecules' such as PKMζ, and the primary site for LTP expression (pre- and/or post-synaptic?). Throughout, I reflect on my life in science, how science is done and what drives it. The reflections are quite personal and I admit to mixed feelings about broadcasting them.
Collapse
Affiliation(s)
- T. Lømo
- Institute of Basic Medical Sciences; University of Oslo; Oslo Norway
| |
Collapse
|
35
|
Mitochondrial Ultrastructure Is Coupled to Synaptic Performance at Axonal Release Sites. eNeuro 2018; 5:eN-NWR-0390-17. [PMID: 29383328 PMCID: PMC5788698 DOI: 10.1523/eneuro.0390-17.2018] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/28/2017] [Accepted: 01/06/2018] [Indexed: 01/08/2023] Open
Abstract
Mitochondrial function in neurons is tightly linked with metabolic and signaling mechanisms that ultimately determine neuronal performance. The subcellular distribution of these organelles is dynamically regulated as they are directed to axonal release sites on demand, but whether mitochondrial internal ultrastructure and molecular properties would reflect the actual performance requirements in a synapse-specific manner, remains to be established. Here, we examined performance-determining ultrastructural features of presynaptic mitochondria in GABAergic and glutamatergic axons of mice and human. Using electron-tomography and super-resolution microscopy we found, that these features were coupled to synaptic strength: mitochondria in boutons with high synaptic activity exhibited an ultrastructure optimized for high rate metabolism and contained higher levels of the respiratory chain protein cytochrome-c (CytC) than mitochondria in boutons with lower activity. The strong, cell type-independent correlation between mitochondrial ultrastructure, molecular fingerprints and synaptic performance suggests that changes in synaptic activity could trigger ultrastructural plasticity of presynaptic mitochondria, likely to adjust their performance to the actual metabolic demand.
Collapse
|
36
|
Assembly of Excitatory Synapses in the Absence of Glutamatergic Neurotransmission. Neuron 2017; 94:312-321.e3. [PMID: 28426966 DOI: 10.1016/j.neuron.2017.03.047] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 02/26/2017] [Accepted: 03/30/2017] [Indexed: 11/22/2022]
Abstract
Synaptic excitation mediates a broad spectrum of structural changes in neural circuits across the brain. Here, we examine the morphologies, wiring, and architectures of single synapses of projection neurons in the murine hippocampus that developed in virtually complete absence of vesicular glutamate release. While these neurons had smaller dendritic trees and/or formed fewer contacts in specific hippocampal subfields, their stereotyped connectivity was largely preserved. Furthermore, loss of release did not disrupt the morphogenesis of presynaptic terminals and dendritic spines, suggesting that glutamatergic neurotransmission is unnecessary for synapse assembly and maintenance. These results underscore the instructive role of intrinsic mechanisms in synapse formation.
Collapse
|
37
|
Loprinzi PD, Edwards MK, Frith E. Potential avenues for exercise to activate episodic memory-related pathways: a narrative review. Eur J Neurosci 2017; 46:2067-2077. [PMID: 28700099 DOI: 10.1111/ejn.13644] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/06/2017] [Accepted: 07/07/2017] [Indexed: 12/13/2022]
Abstract
Memory function plays an important role in activities of daily living, and consequently, quality and quantity of life. In this narrative review, we discuss the anatomical components of episodic memory, including the structure of the hippocampus and the routes of communication to and from this structure. We also highlight cellular traces of memory, such as the engram cell and pathway. To provide etiological insight, the biological mechanisms of episodic memory are discussed, including factors subserving memory encoding (e.g., cognitive attention, neuroelectrical indices), consolidation (i.e., synaptic and brain systems level), and retrieval (e.g., availability of cues, context-dependent, state-dependent, and cognitive processing). Central to this manuscript, we highlight how exercise may influence each of these aforementioned parameters (e.g., exercise-induced hippocampal growth, synaptic plasticity, and cue retrieval) and then discuss the implications of these findings to enhance and preserve memory function. Collectively, this narrative review briefly summarizes potential mechanisms of episodic memory, and how exercise may activate these mechanistic pathways.
Collapse
Affiliation(s)
- Paul D Loprinzi
- Jackson Heart Study Vanguard Center at Oxford, Physical Activity Epidemiology Laboratory, Exercise Psychology Laboratory, Department of Health, Exercise Science and Recreation Management, The University of Mississippi, University, MS, USA
| | - Meghan K Edwards
- Physical Activity Epidemiology Laboratory, Exercise Psychology Laboratory, Department of Health, Exercise Science and Recreation Management, The University of Mississippi, University, MS, USA
| | - Emily Frith
- Physical Activity Epidemiology Laboratory, Exercise Psychology Laboratory, Department of Health, Exercise Science and Recreation Management, The University of Mississippi, University, MS, USA
| |
Collapse
|
38
|
Fakira AK, Massaly N, Cohensedgh O, Berman A, Morón JA. Morphine-Associated Contextual Cues Induce Structural Plasticity in Hippocampal CA1 Pyramidal Neurons. Neuropsychopharmacology 2016; 41:2668-78. [PMID: 27170097 PMCID: PMC5026734 DOI: 10.1038/npp.2016.69] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 04/29/2016] [Accepted: 05/03/2016] [Indexed: 01/08/2023]
Abstract
In people with a prior history of opioid misuse, cues associated with previous drug intake can trigger relapse even after years of abstinence. Examining the processes that lead to the formation and maintenance of the memories between cues/context and the opioid may help to discover new therapeutic candidates to treat drug-seeking behavior. The hippocampus is a brain region essential for learning and memory, which has been involved in the mechanisms underlying opioid cravings. The formation of memories and associations are thought to be dependent on synaptic strengthening associated with structural plasticity of dendritic spines. Here, we assess how dendritic spines in the CA1 region of the hippocampus are affected by morphine-conditioning training. Our results show that morphine pairing with environmental cues (ie, the conditioned place preference (CPP) apparatus) triggers a significant decrease in the number of thin dendritic spines in the hippocampus. Interestingly, this effect was observed regardless of the expression of a conditioned response when mice were trained using an unpaired morphine CPP design and was absent when morphine was administered in the home cage. To investigate the mechanism underlying this structural plasticity, we examined the role of Rho GTPase in dendritic spine remodeling. We found that synaptic expression of RhoA increased with morphine conditioning and blocking RhoA signaling prevented the expression of morphine-induced CPP. Our findings uncover novel mechanisms in response to morphine-associated environmental cues and the underlying alterations in spine plasticity.
Collapse
Affiliation(s)
- Amanda K Fakira
- Department of Anesthesiology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Nicolas Massaly
- Department of Anesthesiology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Omid Cohensedgh
- Department of Anesthesiology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Alexandra Berman
- Department of Anesthesiology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA
| | - Jose A Morón
- Department of Anesthesiology, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY, USA,Department of Anesthesiology, Washington University School of Medicine, Washington University Pain Center, St Louis, MO 63110, USA, Tel: +1 314 362 0078 or +1 314 362 8565, E-mail:
| |
Collapse
|
39
|
Dzyubenko E, Rozenberg A, Hermann DM, Faissner A. Colocalization of synapse marker proteins evaluated by STED-microscopy reveals patterns of neuronal synapse distribution in vitro. J Neurosci Methods 2016; 273:149-159. [PMID: 27615741 DOI: 10.1016/j.jneumeth.2016.09.001] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 08/30/2016] [Accepted: 09/07/2016] [Indexed: 01/08/2023]
Abstract
BACKGROUND Quantification of synapses and their morphological analysis are extensively used in network development and connectivity studies, drug screening and other areas of neuroscience. Thus, a number of quantitative approaches were introduced so far. However, most of the available methods are highly tailored to specific applications and have limitations for widespread use. NEW METHOD We present a new plugin for the open-source software ImageJ to provide a modifiable, high-throughput and easy to use method for synaptic puncta analysis. Our approach is based on colocalization of pre- and postsynaptic protein markers. Structurally completed glutamatergic and GABAergic synapses were identified by VGLUT1-PSD95 and VGAT-gephyrin colocalization, respectively. By combining conventional confocal microscopy with stimulated emission depletion (STED) imaging, we propose a method to quantify the number of scaffolding protein clusters, recruited to a single postsynaptic density. RESULTS In a proof-of-concept study, we reveal the differential distribution of glutamatergic and GABAergic synapse density with reference to perineuronal net (PNN) expression. Using super-resolution STED imaging, we demonstrate that postsynaptic puncta of completed synapses are composed of significantly more protein clusters, compared to uncompleted synapses. COMPARISON WITH EXISTING METHODS Our Synapse Counter plugin for ImageJ offers a rapid and unbiased research tool for a broad spectrum of neuroscientists. The proposed method of synaptic protein clusters quantification exploits super-resolution imaging to provide a comprehensive approach to the analysis of postsynaptic density composition. CONCLUSIONS Our results strongly substantiate the benefits of colocalization-based synapse detection.
Collapse
Affiliation(s)
- Egor Dzyubenko
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Germany; International School of Neuroscience (IGSN), Ruhr-University Bochum, Germany
| | - Andrey Rozenberg
- Department of Animal Ecology, Evolution and Biodiversity, Ruhr-University Bochum, Germany
| | - Dirk M Hermann
- Department of Neurology, University Hospital Essen, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Germany.
| |
Collapse
|
40
|
Mullins C, Fishell G, Tsien RW. Unifying Views of Autism Spectrum Disorders: A Consideration of Autoregulatory Feedback Loops. Neuron 2016; 89:1131-1156. [PMID: 26985722 DOI: 10.1016/j.neuron.2016.02.017] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2016] [Indexed: 12/31/2022]
Abstract
Understanding the mechanisms underlying autism spectrum disorders (ASDs) is a challenging goal. Here we review recent progress on several fronts, including genetics, proteomics, biochemistry, and electrophysiology, that raise motivation for forming a viable pathophysiological hypothesis. In place of a traditionally unidirectional progression, we put forward a framework that extends homeostatic hypotheses by explicitly emphasizing autoregulatory feedback loops and known synaptic biology. The regulated biological feature can be neuronal electrical activity, the collective strength of synapses onto a dendritic branch, the local concentration of a signaling molecule, or the relative strengths of synaptic excitation and inhibition. The sensor of the biological variable (which we have termed the homeostat) engages mechanisms that operate as negative feedback elements to keep the biological variable tightly confined. We categorize known ASD-associated gene products according to their roles in such feedback loops and provide detailed commentary for exemplar genes within each module.
Collapse
Affiliation(s)
- Caitlin Mullins
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA
| | - Gord Fishell
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA
| | - Richard W Tsien
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA.
| |
Collapse
|
41
|
Persistent Structural Plasticity Optimizes Sensory Information Processing in the Olfactory Bulb. Neuron 2016; 91:384-96. [PMID: 27373833 DOI: 10.1016/j.neuron.2016.06.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 04/14/2016] [Accepted: 05/19/2016] [Indexed: 11/23/2022]
Abstract
In the mammalian brain, the anatomical structure of neural circuits changes little during adulthood. As a result, adult learning and memory are thought to result from specific changes in synaptic strength. A possible exception is the olfactory bulb (OB), where activity guides interneuron turnover throughout adulthood. These adult-born granule cell (GC) interneurons form new GABAergic synapses that have little synaptic strength plasticity. In the face of persistent neuronal and synaptic turnover, how does the OB balance flexibility, as is required for adapting to changing sensory environments, with perceptual stability? Here we show that high dendritic spine turnover is a universal feature of GCs, regardless of their developmental origin and age. We find matching dynamics among postsynaptic sites on the principal neurons receiving the new synaptic inputs. We further demonstrate in silico that this coordinated structural plasticity is consistent with stable, yet flexible, decorrelated sensory representations. Together, our study reveals that persistent, coordinated synaptic structural plasticity between interneurons and principal neurons is a major mode of functional plasticity in the OB.
Collapse
|
42
|
Poo MM, Pignatelli M, Ryan TJ, Tonegawa S, Bonhoeffer T, Martin KC, Rudenko A, Tsai LH, Tsien RW, Fishell G, Mullins C, Gonçalves JT, Shtrahman M, Johnston ST, Gage FH, Dan Y, Long J, Buzsáki G, Stevens C. What is memory? The present state of the engram. BMC Biol 2016; 14:40. [PMID: 27197636 PMCID: PMC4874022 DOI: 10.1186/s12915-016-0261-6] [Citation(s) in RCA: 209] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The mechanism of memory remains one of the great unsolved problems of biology. Grappling with the question more than a hundred years ago, the German zoologist Richard Semon formulated the concept of the engram, lasting connections in the brain that result from simultaneous “excitations”, whose precise physical nature and consequences were out of reach of the biology of his day. Neuroscientists now have the knowledge and tools to tackle this question, however, and this Forum brings together leading contemporary views on the mechanisms of memory and what the engram means today.
Collapse
Affiliation(s)
- Mu-Ming Poo
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China.
| | - Michele Pignatelli
- RIKEN-MIT Center for Neural Circuit Genetics at the Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Tomás J Ryan
- RIKEN-MIT Center for Neural Circuit Genetics at the Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.,Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Susumu Tonegawa
- RIKEN-MIT Center for Neural Circuit Genetics at the Picower Institute for Learning and Memory, Department of Biology and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.,Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | | | - Kelsey C Martin
- Department of Biological Chemistry and Department of Psychiatry and Biobehavioral Studies, David Geffen School of Medicine, BSRB 390B, 615 Charles E. Young Dr. South, University of California, Los Angeles, CA, 90095, USA
| | - Andrii Rudenko
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Li-Huei Tsai
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Richard W Tsien
- The Neuroscience Institute, School of Medicine and Center for Neural Science, New York University, New York, NY, 10016, USA
| | - Gord Fishell
- The Neuroscience Institute, School of Medicine and Center for Neural Science, New York University, New York, NY, 10016, USA
| | - Caitlin Mullins
- The Neuroscience Institute, School of Medicine and Center for Neural Science, New York University, New York, NY, 10016, USA
| | - J Tiago Gonçalves
- Salk Institute for Biological Studies, Laboratory of Genetics, 10010 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Matthew Shtrahman
- Salk Institute for Biological Studies, Laboratory of Genetics, 10010 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Stephen T Johnston
- Salk Institute for Biological Studies, Laboratory of Genetics, 10010 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Fred H Gage
- Salk Institute for Biological Studies, Laboratory of Genetics, 10010 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Yang Dan
- HHMI, Department of Molecular and Cell Biology, University of California, Berkeley, USA
| | - John Long
- The Neuroscience Institute, School of Medicine and Center for Neural Science, New York University, New York, NY, 10016, USA
| | - György Buzsáki
- The Neuroscience Institute, School of Medicine and Center for Neural Science, New York University, New York, NY, 10016, USA
| | - Charles Stevens
- Salk Institute for Biological Studies, Laboratory of Genetics, 10010 N. Torrey Pines Road, La Jolla, CA, 92037, USA
| |
Collapse
|
43
|
Glebov OO, Cox S, Humphreys L, Burrone J. Neuronal activity controls transsynaptic geometry. Sci Rep 2016; 6:22703. [PMID: 26951792 PMCID: PMC4782104 DOI: 10.1038/srep22703] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 02/09/2016] [Indexed: 11/13/2022] Open
Abstract
The neuronal synapse is comprised of several distinct zones, including presynaptic vesicle zone (SVZ), active zone (AZ) and postsynaptic density (PSD). While correct relative positioning of these zones is believed to be essential for synaptic function, the mechanisms controlling their mutual localization remain unexplored. Here, we employ high-throughput quantitative confocal imaging, super-resolution and electron microscopy to visualize organization of synaptic subdomains in hippocampal neurons. Silencing of neuronal activity leads to reversible reorganization of the synaptic geometry, resulting in a increased overlap between immunostained AZ and PSD markers; in contrast, the SVZ-AZ spatial coupling is decreased. Bayesian blinking and bleaching (3B) reconstruction reveals that the distance between the AZ-PSD distance is decreased by 30 nm, while electron microscopy shows that the width of the synaptic cleft is decreased by 1.1 nm. Our findings show that multiple aspects of synaptic geometry are dynamically controlled by neuronal activity and suggest mutual repositioning of synaptic components as a potential novel mechanism contributing to the homeostatic forms of synaptic plasticity.
Collapse
Affiliation(s)
- Oleg O. Glebov
- Wolfson Centre for Age-Related Diseases, King’s College London, London SE1 1UL, UK
- Department of Developmental Neurobiology, King’s College London, London SE1 1UL, UK
| | - Susan Cox
- Randall Division of Cell and Molecular Biophysics, King’s College London, London SE1 1UL, UK
| | - Lawrence Humphreys
- Department of Developmental Neurobiology, King’s College London, London SE1 1UL, UK
| | - Juan Burrone
- Department of Developmental Neurobiology, King’s College London, London SE1 1UL, UK
| |
Collapse
|
44
|
Arganda-Carreras I, Turaga SC, Berger DR, Cireşan D, Giusti A, Gambardella LM, Schmidhuber J, Laptev D, Dwivedi S, Buhmann JM, Liu T, Seyedhosseini M, Tasdizen T, Kamentsky L, Burget R, Uher V, Tan X, Sun C, Pham TD, Bas E, Uzunbas MG, Cardona A, Schindelin J, Seung HS. Crowdsourcing the creation of image segmentation algorithms for connectomics. Front Neuroanat 2015; 9:142. [PMID: 26594156 PMCID: PMC4633678 DOI: 10.3389/fnana.2015.00142] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 10/19/2015] [Indexed: 11/13/2022] Open
Abstract
To stimulate progress in automating the reconstruction of neural circuits, we organized the first international challenge on 2D segmentation of electron microscopic (EM) images of the brain. Participants submitted boundary maps predicted for a test set of images, and were scored based on their agreement with a consensus of human expert annotations. The winning team had no prior experience with EM images, and employed a convolutional network. This “deep learning” approach has since become accepted as a standard for segmentation of EM images. The challenge has continued to accept submissions, and the best so far has resulted from cooperation between two teams. The challenge has probably saturated, as algorithms cannot progress beyond limits set by ambiguities inherent in 2D scoring and the size of the test dataset. Retrospective evaluation of the challenge scoring system reveals that it was not sufficiently robust to variations in the widths of neurite borders. We propose a solution to this problem, which should be useful for a future 3D segmentation challenge.
Collapse
Affiliation(s)
- Ignacio Arganda-Carreras
- UMR1318 French National Institute for Agricultural Research-AgroParisTech, French National Institute for Agricultural Research Centre de Versailles-Grignon, Institut Jean-Pierre Bourgin Versailles, France
| | - Srinivas C Turaga
- Howard Hughes Medical Institute, Janelia Research Campus Ashburn, VA, USA
| | - Daniel R Berger
- Center for Brain Science, Harvard University Cambridge, MA, USA
| | - Dan Cireşan
- Swiss AI Lab IDSIA (Dalle Molle Institute for Artificial Intelligence) Universitá Della Svizzera Italiana, Scuola Universitaria Professionale Della Svizzera Italiana Lugano, Switzerland
| | - Alessandro Giusti
- Swiss AI Lab IDSIA (Dalle Molle Institute for Artificial Intelligence) Universitá Della Svizzera Italiana, Scuola Universitaria Professionale Della Svizzera Italiana Lugano, Switzerland
| | - Luca M Gambardella
- Swiss AI Lab IDSIA (Dalle Molle Institute for Artificial Intelligence) Universitá Della Svizzera Italiana, Scuola Universitaria Professionale Della Svizzera Italiana Lugano, Switzerland
| | - Jürgen Schmidhuber
- Swiss AI Lab IDSIA (Dalle Molle Institute for Artificial Intelligence) Universitá Della Svizzera Italiana, Scuola Universitaria Professionale Della Svizzera Italiana Lugano, Switzerland
| | - Dmitry Laptev
- Department of Computer Science, ETH Zurich Zurich, Switzerland
| | - Sarvesh Dwivedi
- Department of Computer Science, ETH Zurich Zurich, Switzerland
| | | | - Ting Liu
- Scientific Computing and Imaging Institute, University of Utah Salt Lake City, UT, USA
| | - Mojtaba Seyedhosseini
- Scientific Computing and Imaging Institute, University of Utah Salt Lake City, UT, USA
| | - Tolga Tasdizen
- Scientific Computing and Imaging Institute, University of Utah Salt Lake City, UT, USA
| | - Lee Kamentsky
- Imaging Platform, Broad Institute Cambridge, MA, USA
| | - Radim Burget
- Department of Telecommunications, Faculty of Electrical Engineering and Communication, Brno University of Technology Brno, Czech Republic
| | - Vaclav Uher
- Department of Telecommunications, Faculty of Electrical Engineering and Communication, Brno University of Technology Brno, Czech Republic
| | - Xiao Tan
- School of Engineering and Information Technology, University of New South Wales Canberra, ACT, Australia
| | - Changming Sun
- Digital Productivity Flagship, Commonwealth Scientific and Industrial Research Organisation North Ryde, NSW, Australia
| | - Tuan D Pham
- Department of Biomedical Engineering, The Institute of Technology, Linkoping University Linkoping, Sweden
| | - Erhan Bas
- Howard Hughes Medical Institute, Janelia Research Campus Ashburn, VA, USA
| | - Mustafa G Uzunbas
- Computer Science Department, Rutgers University New Brunswick, NJ, USA
| | - Albert Cardona
- Howard Hughes Medical Institute, Janelia Research Campus Ashburn, VA, USA
| | - Johannes Schindelin
- Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison Madison, WI, USA
| | - H Sebastian Seung
- Princeton Neuroscience Institute and Computer Science Department, Princeton University Princeton, NJ, USA
| |
Collapse
|
45
|
Structural Components of Synaptic Plasticity and Memory Consolidation. Cold Spring Harb Perspect Biol 2015; 7:a021758. [PMID: 26134321 DOI: 10.1101/cshperspect.a021758] [Citation(s) in RCA: 243] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Consolidation of implicit memory in the invertebrate Aplysia and explicit memory in the mammalian hippocampus are associated with remodeling and growth of preexisting synapses and the formation of new synapses. Here, we compare and contrast structural components of the synaptic plasticity that underlies these two distinct forms of memory. In both cases, the structural changes involve time-dependent processes. Thus, some modifications are transient and may contribute to early formative stages of long-term memory, whereas others are more stable, longer lasting, and likely to confer persistence to memory storage. In addition, we explore the possibility that trans-synaptic signaling mechanisms governing de novo synapse formation during development can be reused in the adult for the purposes of structural synaptic plasticity and memory storage. Finally, we discuss how these mechanisms set in motion structural rearrangements that prepare a synapse to strengthen the same memory and, perhaps, to allow it to take part in other memories as a basis for understanding how their anatomical representation results in the enhanced expression and storage of memories in the brain.
Collapse
|
46
|
Garcia-Alvarez G, Lu B, Yap KAF, Wong LC, Thevathasan JV, Lim L, Ji F, Tan KW, Mancuso JJ, Tang W, Poon SY, Augustine GJ, Fivaz M. STIM2 regulates PKA-dependent phosphorylation and trafficking of AMPARs. Mol Biol Cell 2015; 26:1141-59. [PMID: 25609091 PMCID: PMC4357513 DOI: 10.1091/mbc.e14-07-1222] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
STIMs (STIM1 and STIM2 in mammals) are transmembrane proteins that reside in the endoplasmic reticulum and regulate store-operated Ca2+ entry. STIM2 mediates cAMP/PKA-dependent phosphorylation of the AMPA receptor subunit GluA1 in excitatory neurons. In addition, STIM2 promotes cAMP-dependent surface delivery of GluA1. STIMs (STIM1 and STIM2 in mammals) are transmembrane proteins that reside in the endoplasmic reticulum (ER) and regulate store-operated Ca2+ entry (SOCE). The function of STIMs in the brain is only beginning to be explored, and the relevance of SOCE in nerve cells is being debated. Here we identify STIM2 as a central organizer of excitatory synapses. STIM2, but not its paralogue STIM1, influences the formation of dendritic spines and shapes basal synaptic transmission in excitatory neurons. We further demonstrate that STIM2 is essential for cAMP/PKA-dependent phosphorylation of the AMPA receptor (AMPAR) subunit GluA1. cAMP triggers rapid migration of STIM2 to ER–plasma membrane (PM) contact sites, enhances recruitment of GluA1 to these ER-PM junctions, and promotes localization of STIM2 in dendritic spines. Both biochemical and imaging data suggest that STIM2 regulates GluA1 phosphorylation by coupling PKA to the AMPAR in a SOCE-independent manner. Consistent with a central role of STIM2 in regulating AMPAR phosphorylation, STIM2 promotes cAMP-dependent surface delivery of GluA1 through combined effects on exocytosis and endocytosis. Collectively our results point to a unique mechanism of synaptic plasticity driven by dynamic assembly of a STIM2 signaling complex at ER-PM contact sites.
Collapse
Affiliation(s)
- Gisela Garcia-Alvarez
- Program in Neuroscience and Behavioral Disorders, DUKE-NUS Graduate Medical School, Singapore 169857
| | - Bo Lu
- Program in Neuroscience and Behavioral Disorders, DUKE-NUS Graduate Medical School, Singapore 169857
| | - Kenrick An Fu Yap
- Program in Neuroscience and Behavioral Disorders, DUKE-NUS Graduate Medical School, Singapore 169857
| | - Loo Chin Wong
- Program in Neuroscience and Behavioral Disorders, DUKE-NUS Graduate Medical School, Singapore 169857
| | - Jervis Vermal Thevathasan
- Program in Neuroscience and Behavioral Disorders, DUKE-NUS Graduate Medical School, Singapore 169857
| | - Lynette Lim
- Program in Neuroscience and Behavioral Disorders, DUKE-NUS Graduate Medical School, Singapore 169857
| | - Fang Ji
- Program in Neuroscience and Behavioral Disorders, DUKE-NUS Graduate Medical School, Singapore 169857
| | - Kia Wee Tan
- Program in Neuroscience and Behavioral Disorders, DUKE-NUS Graduate Medical School, Singapore 169857
| | - James J Mancuso
- Program in Neuroscience and Behavioral Disorders, DUKE-NUS Graduate Medical School, Singapore 169857
| | - Willcyn Tang
- Program in Neuroscience and Behavioral Disorders, DUKE-NUS Graduate Medical School, Singapore 169857
| | - Shou Yu Poon
- Program in Neuroscience and Behavioral Disorders, DUKE-NUS Graduate Medical School, Singapore 169857
| | - George J Augustine
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 637553 Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul 136-791, Republic of Korea
| | - Marc Fivaz
- Program in Neuroscience and Behavioral Disorders, DUKE-NUS Graduate Medical School, Singapore 169857 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
| |
Collapse
|
47
|
Adrian M, Kusters R, Wierenga CJ, Storm C, Hoogenraad CC, Kapitein LC. Barriers in the brain: resolving dendritic spine morphology and compartmentalization. Front Neuroanat 2014; 8:142. [PMID: 25538570 PMCID: PMC4255500 DOI: 10.3389/fnana.2014.00142] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 11/13/2014] [Indexed: 11/23/2022] Open
Abstract
Dendritic spines are micron-sized protrusions that harbor the majority of excitatory synapses in the central nervous system. The head of the spine is connected to the dendritic shaft by a 50-400 nm thin membrane tube, called the spine neck, which has been hypothesized to confine biochemical and electric signals within the spine compartment. Such compartmentalization could minimize interspinal crosstalk and thereby support spine-specific synapse plasticity. However, to what extent compartmentalization is governed by spine morphology, and in particular the diameter of the spine neck, has remained unresolved. Here, we review recent advances in tool development - both experimental and theoretical - that facilitate studying the role of the spine neck in compartmentalization. Special emphasis is given to recent advances in microscopy methods and quantitative modeling applications as we discuss compartmentalization of biochemical signals, membrane receptors and electrical signals in spines. Multidisciplinary approaches should help to answer how dendritic spine architecture affects the cellular and molecular processes required for synapse maintenance and modulation.
Collapse
Affiliation(s)
- Max Adrian
- Cell Biology, Department of Biology, Faculty of Science, Utrecht UniversityUtrecht, Netherlands
| | - Remy Kusters
- Department of Applied Physics, Eindhoven University of TechnologyEindhoven, Netherlands
| | - Corette J. Wierenga
- Cell Biology, Department of Biology, Faculty of Science, Utrecht UniversityUtrecht, Netherlands
| | - Cornelis Storm
- Department of Applied Physics, Eindhoven University of TechnologyEindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of TechnologyEindhoven, Netherlands
| | - Casper C. Hoogenraad
- Cell Biology, Department of Biology, Faculty of Science, Utrecht UniversityUtrecht, Netherlands
| | - Lukas C. Kapitein
- Cell Biology, Department of Biology, Faculty of Science, Utrecht UniversityUtrecht, Netherlands
| |
Collapse
|
48
|
Bell ME, Bourne JN, Chirillo MA, Mendenhall JM, Kuwajima M, Harris KM. Dynamics of nascent and active zone ultrastructure as synapses enlarge during long-term potentiation in mature hippocampus. J Comp Neurol 2014; 522:3861-84. [PMID: 25043676 PMCID: PMC4167938 DOI: 10.1002/cne.23646] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 06/23/2014] [Accepted: 06/30/2014] [Indexed: 11/08/2022]
Abstract
Nascent zones and active zones are adjacent synaptic regions that share a postsynaptic density, but nascent zones lack the presynaptic vesicles found at active zones. Here dendritic spine synapses were reconstructed through serial section electron microscopy (3DEM) and EM tomography to investigate nascent zone dynamics during long-term potentiation (LTP) in mature rat hippocampus. LTP was induced with theta-burst stimulation, and comparisons were made with control stimulation in the same hippocampal slices at 5 minutes, 30 minutes, and 2 hours post-induction and to perfusion-fixed hippocampus in vivo. Nascent zones were present at the edges of ∼35% of synapses in perfusion-fixed hippocampus and as many as ∼50% of synapses in some hippocampal slice conditions. By 5 minutes, small dense-core vesicles known to transport active zone proteins moved into more presynaptic boutons. By 30 minutes, nascent zone area decreased, without significant change in synapse area, suggesting that presynaptic vesicles were recruited to preexisting nascent zones. By 2 hours, both nascent and active zones were enlarged. Immunogold labeling revealed glutamate receptors in nascent zones; however, average distances from nascent zones to docked presynaptic vesicles ranged from 170 ± 5 nm in perfusion-fixed hippocampus to 251 ± 4 nm at enlarged synapses by 2 hours during LTP. Prior stochastic modeling suggests that decrease in glutamate concentration reduces the probability of glutamate receptor activation from 0.4 at the center of release to 0.1 just 200 nm away. Thus, conversion of nascent zones to functional active zones likely requires the recruitment of presynaptic vesicles during LTP.
Collapse
Affiliation(s)
- Maria Elizabeth Bell
- Center for Learning and Memory, Department of Neuroscience, Institute for Neuroscience, University of Texas, Austin, TX 78712
| | - Jennifer N. Bourne
- Center for Learning and Memory, Department of Neuroscience, Institute for Neuroscience, University of Texas, Austin, TX 78712
| | - Michael A. Chirillo
- Center for Learning and Memory, Department of Neuroscience, Institute for Neuroscience, University of Texas, Austin, TX 78712
- The University of Texas Medical School, Houston, TX 77030
| | - John M. Mendenhall
- Center for Learning and Memory, Department of Neuroscience, Institute for Neuroscience, University of Texas, Austin, TX 78712
| | - Masaaki Kuwajima
- Center for Learning and Memory, Department of Neuroscience, Institute for Neuroscience, University of Texas, Austin, TX 78712
| | - Kristen M. Harris
- Center for Learning and Memory, Department of Neuroscience, Institute for Neuroscience, University of Texas, Austin, TX 78712
| |
Collapse
|
49
|
Cao G, Harris KM. Augmenting saturated LTP by broadly spaced episodes of theta-burst stimulation in hippocampal area CA1 of adult rats and mice. J Neurophysiol 2014; 112:1916-24. [PMID: 25057146 DOI: 10.1152/jn.00297.2014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hippocampal long-term potentiation (LTP) is a model system for studying cellular mechanisms of learning and memory. Recent interest in mechanisms underlying the advantage of spaced over massed learning has prompted investigation into the effects of distributed episodes of LTP induction. The amount of LTP induced in hippocampal area CA1 by one train (1T) of theta-burst stimulation (TBS) in young Sprague-Dawley rats was further enhanced by additional bouts of 1T given at 1-h intervals. However, in young Long-Evans (LE) rats, 1T did not initially saturate LTP. Instead, a stronger LTP induction paradigm using eight trains of TBS (8T) induced saturated LTP in hippocampal slices from both young and adult LE rats as well as adult mice. The saturated LTP induced by 8T could be augmented by another episode of 8T following an interval of at least 90 min. The success rate across animals and slices in augmenting LTP by an additional episode of 8T increased significantly with longer intervals between the first and last episodes, ranging from 0% at 30- and 60-min intervals to 13-66% at 90- to 180-min intervals to 90-100% at 240-min intervals. Augmentation above initially saturated LTP was blocked by the N-methyl-D-aspartate (NMDA) glutamate receptor antagonist D-2-amino-5-phosphonovaleric acid (D-APV). These findings suggest that the strength of induction and interval between episodes of TBS, as well as the strain and age of the animal, are important components in the augmentation of LTP.
Collapse
Affiliation(s)
- Guan Cao
- The Center for Learning and Memory, University of Texas at Austin, Austin, Texas
| | - Kristen M Harris
- The Center for Learning and Memory, University of Texas at Austin, Austin, Texas
| |
Collapse
|
50
|
Ueda Y. The Role of Phosphoinositides in Synapse Function. Mol Neurobiol 2014; 50:821-38. [DOI: 10.1007/s12035-014-8768-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Accepted: 06/01/2014] [Indexed: 11/30/2022]
|