1
|
Su X, He J, Liu Y, Liu K, Lei B, Zhong Y. Protocol for labeling and manipulating "home"-activated neurons in mice. STAR Protoc 2025; 6:103704. [PMID: 40121664 PMCID: PMC11981720 DOI: 10.1016/j.xpro.2025.103704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/22/2025] [Accepted: 02/26/2025] [Indexed: 03/25/2025] Open
Abstract
Home-related cues, such as home bedding, influence affective behaviors in mice, including fear relief, anxiety reduction, and defensive aggression. Here, we present a protocol for labeling and manipulating "home"-activated neurons in the lateral hypothalamus (LH) using a Tet-Off inducible system. We describe steps for virus injection, postoperative recovery, and combining c-Fos-driven activity labeling with chemogenetics. This protocol integrates neuronal targeting with behavioral assays to provide insights into the functional role of "home"-activated neurons in home-driven behavioral adaptations. For complete details on the use and execution of this protocol, please refer to Su et al.1.
Collapse
Affiliation(s)
- Xiaoya Su
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, P.R. China; School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; McGovern Institute of Brain Research, Beijing 100084, P.R. China
| | - Junyue He
- School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; McGovern Institute of Brain Research, Beijing 100084, P.R. China; Peking University, Tsinghua University, National Institute Biological Science Joint Graduate Program, Beijing, P.R. China
| | - Yunlong Liu
- Neuroscience and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Keyu Liu
- McGovern Institute of Brain Research, Beijing 100084, P.R. China
| | - Bo Lei
- Beijing Academy of Artificial Intelligence, Beijing 100084, P.R. China.
| | - Yi Zhong
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, P.R. China; School of Life Sciences, Tsinghua University, Beijing 100084, P.R. China; McGovern Institute of Brain Research, Beijing 100084, P.R. China; MOE Key Laboratory of Protein Sciences, Tsinghua University, Beijing 100084, P.R. China.
| |
Collapse
|
2
|
Li Z, Lu W, Yang L, Lai N, Wang Y, Chen Z. Decade of TRAP progress: Insights and future prospects for advancing functional network research in epilepsy. Prog Neurobiol 2025; 244:102707. [PMID: 39725016 DOI: 10.1016/j.pneurobio.2024.102707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/30/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
Targeted Recombination in Active Populations (TRAP) represents an effective and extensively applied technique that has earned significant utilization in neuroscience over the past decade, primarily for identifying and modulating functionally activated neuronal ensembles associated with diverse behaviors. As epilepsy is a neurological disorder characterized by pathological hyper-excitatory networks, TRAP has already been widely applied in epilepsy research. However, the deployment of TRAP in this field remains underexplored, and there is significant potential for further application and development in epilepsy-related investigations. In this review, we embark on a concise examination of the mechanisms behind several TRAP tools, introduce the current applications of TRAP in epilepsy research, and collate the key advantages as well as limitations of TRAP. Furthermore, we sketch out perspectives on potential applications of TRAP in future epilepsy research, grounded in the present landscape and challenges of the field, as well as the ways TRAP has been embraced in other neuroscience domains.
Collapse
Affiliation(s)
- Zhisheng Li
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wangjialu Lu
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lin Yang
- key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Nanxi Lai
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yi Wang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China; key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Zhong Chen
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, China; key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
3
|
Bijoch Ł, Włodkowska U, Kasztelanic R, Pawłowska M, Pysz D, Kaczmarek L, Lapkiewicz R, Buczyński R, Czajkowski R. Novel Design and Application of High-NA Fiber Imaging Bundles for In Vivo Brain Imaging with Two-Photon Scanning Fluorescence Microscopy. ACS APPLIED MATERIALS & INTERFACES 2023; 15:12831-12841. [PMID: 36880640 PMCID: PMC10020965 DOI: 10.1021/acsami.2c22985] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/21/2023] [Indexed: 06/18/2023]
Abstract
Here, we provide experimental verification supporting the use of short-section imaging bundles for two-photon microscopy imaging of the mouse brain. The 8 mm long bundle is made of a pair of heavy-metal oxide glasses with a refractive index contrast of 0.38 to ensure a high numerical aperture NA = 1.15. The bundle is composed of 825 multimode cores, ordered in a hexagonal lattice with a pixel size of 14 μm and a total diameter of 914 μm. We demonstrate successful imaging through custom-made bundles with 14 μm resolution. As the input, we used a 910 nm Ti-sapphire laser with 140 fs pulse and a peak power of 9 × 104 W. The excitation beam and fluorescent image were transferred through the fiber imaging bundle. As test samples, we used 1 μm green fluorescent latex beads, ex vivo hippocampal neurons expressing green fluorescent protein and cortical neurons in vivo expressing the fluorescent reporter GCaMP6s or immediate early gene Fos fluorescent reporter. This system can be used for minimal-invasive in vivo imaging of the cerebral cortex, hippocampus, or deep brain areas as a part of a tabletop system or an implantable setup. It is a low-cost solution, easy to integrate and operate for high-throughput experiments.
Collapse
Affiliation(s)
- Łukasz Bijoch
- BRAINCITY, Nencki Institute of Experimental Biology PAS, Pasteura 3, 02-093 Warszawa, Poland
| | - Urszula Włodkowska
- Nencki
Institute of Experimental Biology PAS, Pasteura 3, 02-093 Warszawa, Poland
| | - Rafał Kasztelanic
- Faculty
of Physics, University of Warsaw, Pasteura 5, 02-093 Warsaw, Poland
- Institute
of Microelectronics and Photonics, Lukasiewicz
Research Network, Al.
Lotników 32/46, 02-668 Warsaw, Poland
| | - Monika Pawłowska
- BRAINCITY, Nencki Institute of Experimental Biology PAS, Pasteura 3, 02-093 Warszawa, Poland
- Faculty
of Physics, University of Warsaw, Pasteura 5, 02-093 Warsaw, Poland
| | - Dariusz Pysz
- Institute
of Microelectronics and Photonics, Lukasiewicz
Research Network, Al.
Lotników 32/46, 02-668 Warsaw, Poland
| | - Leszek Kaczmarek
- BRAINCITY, Nencki Institute of Experimental Biology PAS, Pasteura 3, 02-093 Warszawa, Poland
| | - Radek Lapkiewicz
- Faculty
of Physics, University of Warsaw, Pasteura 5, 02-093 Warsaw, Poland
| | - Ryszard Buczyński
- Faculty
of Physics, University of Warsaw, Pasteura 5, 02-093 Warsaw, Poland
- Institute
of Microelectronics and Photonics, Lukasiewicz
Research Network, Al.
Lotników 32/46, 02-668 Warsaw, Poland
| | - Rafał Czajkowski
- Nencki
Institute of Experimental Biology PAS, Pasteura 3, 02-093 Warszawa, Poland
| |
Collapse
|
4
|
Swanson JL, Chin PS, Romero JM, Srivastava S, Ortiz-Guzman J, Hunt PJ, Arenkiel BR. Advancements in the Quest to Map, Monitor, and Manipulate Neural Circuitry. Front Neural Circuits 2022; 16:886302. [PMID: 35719420 PMCID: PMC9204427 DOI: 10.3389/fncir.2022.886302] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/27/2022] [Indexed: 01/27/2023] Open
Abstract
Neural circuits and the cells that comprise them represent the functional units of the brain. Circuits relay and process sensory information, maintain homeostasis, drive behaviors, and facilitate cognitive functions such as learning and memory. Creating a functionally-precise map of the mammalian brain requires anatomically tracing neural circuits, monitoring their activity patterns, and manipulating their activity to infer function. Advancements in cell-type-specific genetic tools allow interrogation of neural circuits with increased precision. This review provides a broad overview of recombination-based and activity-driven genetic targeting approaches, contemporary viral tracing strategies, electrophysiological recording methods, newly developed calcium, and voltage indicators, and neurotransmitter/neuropeptide biosensors currently being used to investigate circuit architecture and function. Finally, it discusses methods for acute or chronic manipulation of neural activity, including genetically-targeted cellular ablation, optogenetics, chemogenetics, and over-expression of ion channels. With this ever-evolving genetic toolbox, scientists are continuing to probe neural circuits with increasing resolution, elucidating the structure and function of the incredibly complex mammalian brain.
Collapse
Affiliation(s)
- Jessica L. Swanson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Pey-Shyuan Chin
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Juan M. Romero
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, United States
| | - Snigdha Srivastava
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, United States
| | - Joshua Ortiz-Guzman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
| | - Patrick J. Hunt
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, United States
| | - Benjamin R. Arenkiel
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, United States
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
5
|
Li D, Wang G, Werner R, Xie H, Guan JS, Hilgetag CC. Single Image-Based Vignetting Correction for Improving the Consistency of Neural Activity Analysis in 2-Photon Functional Microscopy. Front Neuroinform 2022; 15:674439. [PMID: 35069164 PMCID: PMC8766855 DOI: 10.3389/fninf.2021.674439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 12/01/2021] [Indexed: 12/04/2022] Open
Abstract
High-resolution functional 2-photon microscopy of neural activity is a cornerstone technique in current neuroscience, enabling, for instance, the image-based analysis of relations of the organization of local neuron populations and their temporal neural activity patterns. Interpreting local image intensity as a direct quantitative measure of neural activity presumes, however, a consistent within- and across-image relationship between the image intensity and neural activity, which may be subject to interference by illumination artifacts. In particular, the so-called vignetting artifact—the decrease of image intensity toward the edges of an image—is, at the moment, widely neglected in the context of functional microscopy analyses of neural activity, but potentially introduces a substantial center-periphery bias of derived functional measures. In the present report, we propose a straightforward protocol for single image-based vignetting correction. Using immediate-early gene-based 2-photon microscopic neural image data of the mouse brain, we show the necessity of correcting both image brightness and contrast to improve within- and across-image intensity consistency and demonstrate the plausibility of the resulting functional data.
Collapse
Affiliation(s)
- Dong Li
- Institute of Computational Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- *Correspondence: Dong Li,
| | - Guangyu Wang
- School of Life Sciences and Technology, ShanghaiTech University, Shanghai, China
| | - René Werner
- Institute of Computational Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Center for Biomedical Artificial Intelligence (bAIome), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hong Xie
- Institute of Photonic Chips, University of Shanghai for Science and Technology, Shanghai, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Ji-Song Guan
- School of Life Sciences and Technology, ShanghaiTech University, Shanghai, China
- Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Claus C. Hilgetag
- Institute of Computational Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Center for Biomedical Artificial Intelligence (bAIome), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Health Sciences, Boston University, Boston, MA, United States
| |
Collapse
|
6
|
Balcerek E, Włodkowska U, Czajkowski R. Retrosplenial cortex in spatial memory: focus on immediate early genes mapping. Mol Brain 2021; 14:172. [PMID: 34863215 PMCID: PMC8642902 DOI: 10.1186/s13041-021-00880-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/10/2021] [Indexed: 11/10/2022] Open
Abstract
The ability to form, retrieve and update autobiographical memories is one of the most fascinating features of human behavior. Spatial memory, the ability to remember the layout of the external environment and to navigate within its boundaries, is closely related to the autobiographical memory domain. It is served by an overlapping brain circuit, centered around the hippocampus (HPC) where the cognitive map index is stored. Apart from the hippocampus, several cortical structures participate in this process. Their relative contribution is a subject of intense research in both humans and animal models. One of the most widely studied regions is the retrosplenial cortex (RSC), an area in the parietal lobe densely interconnected with the hippocampal formation. Several methodological approaches have been established over decades in order to investigate the cortical aspects of memory. One of the most successful techniques is based on the analysis of brain expression patterns of the immediate early genes (IEGs). The common feature of this diverse group of genes is fast upregulation of their mRNA translation upon physiologically relevant stimulus. In the central nervous system they are rapidly triggered by neuronal activity and plasticity during learning. There is a widely accepted consensus that their expression level corresponds to the engagement of individual neurons in the formation of memory trace. Imaging of the IEGs might therefore provide a picture of an emerging memory engram. In this review we present the overview of IEG mapping studies of retrosplenial cortex in rodent models. We begin with classical techniques, immunohistochemical detection of protein and fluorescent in situ hybridization of mRNA. We then proceed to advanced methods where fluorescent genetically encoded IEG reporters are chronically followed in vivo during memory formation. We end with a combination of genetic IEG labelling and optogenetic approach, where the activity of the entire engram is manipulated. We finally present a hypothesis that attempts to unify our current state of knowledge about the function of RSC.
Collapse
Affiliation(s)
- Edyta Balcerek
- Laboratory of Spatial Memory, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093, Warsaw, Poland
| | - Urszula Włodkowska
- Laboratory of Spatial Memory, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093, Warsaw, Poland
| | - Rafał Czajkowski
- Laboratory of Spatial Memory, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093, Warsaw, Poland.
| |
Collapse
|
7
|
Yoon SH, Song WS, Oh SP, Kim YS, Kim MH. The phosphorylation status of eukaryotic elongation factor-2 indicates neural activity in the brain. Mol Brain 2021; 14:142. [PMID: 34526091 PMCID: PMC8442277 DOI: 10.1186/s13041-021-00852-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/04/2021] [Indexed: 11/21/2022] Open
Abstract
Assessment of neural activity in the specific brain area is critical for understanding the circuit mechanisms underlying altered brain function and behaviors. A number of immediate early genes (IEGs) that are rapidly transcribed in neuronal cells in response to synaptic activity have been used as markers for neuronal activity. However, protein detection of IEGs requires translation, and the amount of newly synthesized gene product is usually insufficient to detect using western blotting, limiting their utility in western blot analysis of brain tissues for comparison of basal activity between control and genetically modified animals. Here, we show that the phosphorylation status of eukaryotic elongation factor-2 (eEF2) rapidly changes in response to synaptic and neural activities. Intraperitoneal injections of the GABA A receptor (GABAAR) antagonist picrotoxin and the glycine receptor antagonist brucine rapidly dephosphorylated eEF2. Conversely, potentiation of GABAARs or inhibition of AMPA receptors (AMPARs) induced rapid phosphorylation of eEF2 in both the hippocampus and forebrain of mice. Chemogenetic suppression of hippocampal principal neuron activity promoted eEF2 phosphorylation. Novel context exploration and acute restraint stress rapidly modified the phosphorylation status of hippocampal eEF2. Furthermore, the hippocampal eEF2 phosphorylation levels under basal conditions were reduced in mice exhibiting epilepsy and abnormally enhanced excitability in CA3 pyramidal neurons. Collectively, the results indicated that eEF2 phosphorylation status is sensitive to neural activity and the ratio of phosphorylated eEF2 to total eEF2 could be a molecular signature for estimating neural activity in a specific brain area.
Collapse
Affiliation(s)
- Sang Ho Yoon
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea.,Neuroscience Research Institute, Seoul National University Medical Research Center, Seoul, 03080, Korea
| | - Woo Seok Song
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea.,Neuroscience Research Institute, Seoul National University Medical Research Center, Seoul, 03080, Korea
| | - Sung Pyo Oh
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Young Sook Kim
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Myoung-Hwan Kim
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Korea. .,Neuroscience Research Institute, Seoul National University Medical Research Center, Seoul, 03080, Korea. .,Seoul National University Bundang Hospital, Seongnam, 13620, Gyeonggi, Korea.
| |
Collapse
|
8
|
Ryan TJ, Ortega-de San Luis C, Pezzoli M, Sen S. Engram cell connectivity: an evolving substrate for information storage. Curr Opin Neurobiol 2021; 67:215-225. [PMID: 33812274 DOI: 10.1016/j.conb.2021.01.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/15/2021] [Accepted: 01/16/2021] [Indexed: 01/02/2023]
Abstract
Understanding memory requires an explanation for how information can be stored in the brain in a stable state. The change in the brain that accounts for a given memory is referred to as an engram. In recent years, the term engram has been operationalized as the cells that are activated by a learning experience, undergoes plasticity, and are sufficient and necessary for memory recall. Using this framework, and a growing toolbox of related experimental techniques, engram manipulation has become a central topic in behavioral, systems, and molecular neuroscience. Recent research on the topic has provided novel insights into the mechanisms of long-term memory storage, and its overlap with instinct. We propose that memory and instinct may be embodied as isomorphic topological structures within the brain's microanatomical circuitry.
Collapse
Affiliation(s)
- Tomás J Ryan
- School of Biochemistry and Immunology and Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, D02 PN40, Ireland; Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC 3052, Australia; Child & Brain Development Program, Canadian Institute for Advanced Research (CIFAR), Toronto, Ontario M5G 1M1, Canada.
| | - Clara Ortega-de San Luis
- School of Biochemistry and Immunology and Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, D02 PN40, Ireland
| | - Maurizio Pezzoli
- School of Biochemistry and Immunology and Trinity College Institute for Neuroscience, Trinity College Dublin, Dublin, D02 PN40, Ireland
| | - Siddhartha Sen
- Centre for Research on Adaptive Nanostructures and Nanodevices and School of Physics, Trinity College Dublin, D02 PN40, Ireland
| |
Collapse
|
9
|
Pléau C, Peret A, Pearlstein E, Scalfati T, Vigier A, Marti G, Michel FJ, Marissal T, Crépel V. Dentate Granule Cells Recruited in the Home Environment Display Distinctive Properties. Front Cell Neurosci 2021; 14:609123. [PMID: 33519383 PMCID: PMC7843370 DOI: 10.3389/fncel.2020.609123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/23/2020] [Indexed: 11/13/2022] Open
Abstract
The dentate granule cells (DGCs) play a crucial role in learning and memory. Many studies have described the role and physiological properties of these sparsely active neurons using different behavioral contexts. However, the morpho-functional features of DGCs recruited in mice maintained in their home cage (without training), considered as a baseline condition, have not yet been established. Using fosGFP transgenic mice, we observed ex vivo that DGCs recruited in animals maintained in the home cage condition are mature neurons that display a longer dendritic tree and lower excitability compared with non-activated cells. The higher GABAA receptor-mediated shunting inhibition contributes to the lower excitability of DGCs activated in the home environment by shifting the input resistance towards lower values. Remarkably, that shunting inhibition is neither observed in non-activated DGCs nor in DGCs activated during training in virtual reality. In short, our results suggest that strong shunting inhibition and reduced excitability could constitute a distinctive neural signature of mature DGCs recruited in the context of the home environment.
Collapse
Affiliation(s)
- Claire Pléau
- INMED, INSERM UMR1249, Aix-Marseille University, Marseille, France
| | - Angélique Peret
- INMED, INSERM UMR1249, Aix-Marseille University, Marseille, France
| | | | - Thomas Scalfati
- INMED, INSERM UMR1249, Aix-Marseille University, Marseille, France
| | - Alexandre Vigier
- INMED, INSERM UMR1249, Aix-Marseille University, Marseille, France
| | | | | | - Thomas Marissal
- INMED, INSERM UMR1249, Aix-Marseille University, Marseille, France
| | - Valérie Crépel
- INMED, INSERM UMR1249, Aix-Marseille University, Marseille, France
| |
Collapse
|
10
|
Hippocampal neurons with stable excitatory connectivity become part of neuronal representations. PLoS Biol 2020; 18:e3000928. [PMID: 33141818 PMCID: PMC7665705 DOI: 10.1371/journal.pbio.3000928] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 11/13/2020] [Accepted: 09/24/2020] [Indexed: 12/22/2022] Open
Abstract
Experiences are represented in the brain by patterns of neuronal activity. Ensembles of neurons representing experience undergo activity-dependent plasticity and are important for learning and recall. They are thus considered cellular engrams of memory. Yet, the cellular events that bias neurons to become part of a neuronal representation are largely unknown. In rodents, turnover of structural connectivity has been proposed to underlie the turnover of neuronal representations and also to be a cellular mechanism defining the time duration for which memories are stored in the hippocampus. If these hypotheses are true, structural dynamics of connectivity should be involved in the formation of neuronal representations and concurrently important for learning and recall. To tackle these questions, we used deep-brain 2-photon (2P) time-lapse imaging in transgenic mice in which neurons expressing the Immediate Early Gene (IEG) Arc (activity-regulated cytoskeleton-associated protein) could be permanently labeled during a specific time window. This enabled us to investigate the dynamics of excitatory synaptic connectivity—using dendritic spines as proxies—of hippocampal CA1 (cornu ammonis 1) pyramidal neurons (PNs) becoming part of neuronal representations exploiting Arc as an indicator of being part of neuronal representations. We discovered that neurons that will prospectively express Arc have slower turnover of synaptic connectivity, thus suggesting that synaptic stability prior to experience can bias neurons to become part of representations or possibly engrams. We also found a negative correlation between stability of structural synaptic connectivity and the ability to recall features of a hippocampal-dependent memory, which suggests that faster structural turnover in hippocampal CA1 might be functional for memory. The cellular events that bias neurons to become part of neuronal representations and engrams are largely unknown. This study of the dynamics of excitatory synaptic connectivity of CA1 hippocampal neurons expressing the Immediate Early Gene Arc reveals that synaptic stability can bias neurons to become part of representations and that faster structural turnover in dorsal hippocampal CA1 might be functional for memory.
Collapse
|
11
|
Simbriger K, Amorim IS, Lach G, Chalkiadaki K, Kouloulia S, Jafarnejad SM, Khoutorsky A, Gkogkas CG. Uncovering memory-related gene expression in contextual fear conditioning using ribosome profiling. Prog Neurobiol 2020; 197:101903. [PMID: 32860876 PMCID: PMC7859833 DOI: 10.1016/j.pneurobio.2020.101903] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 06/30/2020] [Accepted: 08/23/2020] [Indexed: 11/14/2022]
Abstract
Contextual fear conditioning (CFC) in rodents is the most widely used behavioural paradigm in neuroscience research to elucidate the neurobiological mechanisms underlying learning and memory. It is based on the pairing of an aversive unconditioned stimulus (US; e.g. mild footshock) with a neutral conditioned stimulus (CS; e.g. context of the test chamber) in order to acquire associative long-term memory (LTM), which persists for days and even months. Using genome-wide analysis, several studies have generated lists of genes modulated in response to CFC in an attempt to identify the “memory genes”, which orchestrate memory formation. Yet, most studies use naïve animals as a baseline for assessing gene-expression changes, while only few studies have examined the effect of the US alone, without pairing to context, using genome-wide analysis of gene-expression. Herein, using the ribosome profiling methodology, we show that in male mice an immediate shock, which does not lead to LTM formation, elicits pervasive translational and transcriptional changes in the expression of Immediate Early Genes (IEGs) in dorsal hippocampus (such as Fos and Arc), a fact which has been disregarded by the majority of CFC studies. By removing the effect of the immediate shock, we identify and validate a new set of genes, which are translationally and transcriptionally responsive to the association of context-to-footshock in CFC, and thus constitute salient “memory genes”.
Collapse
Affiliation(s)
- Konstanze Simbriger
- Centre for Discovery Brain Sciences, University of Edinburgh and Patrick Wild Centre and Simons Initiative for the Developing Brain, University of Edinburgh, EH8 9XD, Edinburgh, Scotland, UK
| | - Inês S Amorim
- Centre for Discovery Brain Sciences, University of Edinburgh and Patrick Wild Centre and Simons Initiative for the Developing Brain, University of Edinburgh, EH8 9XD, Edinburgh, Scotland, UK
| | - Gilliard Lach
- Centre for Discovery Brain Sciences, University of Edinburgh and Patrick Wild Centre and Simons Initiative for the Developing Brain, University of Edinburgh, EH8 9XD, Edinburgh, Scotland, UK
| | - Kleanthi Chalkiadaki
- Centre for Discovery Brain Sciences, University of Edinburgh and Patrick Wild Centre and Simons Initiative for the Developing Brain, University of Edinburgh, EH8 9XD, Edinburgh, Scotland, UK
| | - Stella Kouloulia
- Centre for Discovery Brain Sciences, University of Edinburgh and Patrick Wild Centre and Simons Initiative for the Developing Brain, University of Edinburgh, EH8 9XD, Edinburgh, Scotland, UK
| | - Seyed Mehdi Jafarnejad
- Patrick G. Johnston Centre for Cancer Research and Cell Biology, The Queen's University of Belfast, BT9 7AE Belfast, Northern Ireland, UK.
| | - Arkady Khoutorsky
- Department of Anesthesia, Faculty of Dentistry and Alan Edwards Centre for Research on Pain, McGill University, H3A 0G1, Montréal, QC, Canada.
| | - Christos G Gkogkas
- Centre for Discovery Brain Sciences, University of Edinburgh and Patrick Wild Centre and Simons Initiative for the Developing Brain, University of Edinburgh, EH8 9XD, Edinburgh, Scotland, UK.
| |
Collapse
|
12
|
Metasuk A, Kitiyanant N, Chetsawang B. Expression of nano-ferritin in neuronal cells encompassed by minimal Arc promoter system. Biochem Biophys Res Commun 2020; 526:574-579. [PMID: 32247609 DOI: 10.1016/j.bbrc.2020.03.105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 03/18/2020] [Indexed: 11/18/2022]
Abstract
Genetic engineering for neuronal cell activity labeling and neuronal cell activity modulation are invaluable for elucidating the underlying characteristics of the brain and neurons. In this study, ferritin fusion protein (FFP) was combined with Tet expression construct under a modified immediate-early gene (IEG) Arc/Arg3.1 promoter so-called SARE-ArcMin. This expression system is a neuronal activity-dependent expression module for nano-ferritin, a radio/magnetic wave-sensitive protein well-accepted as a potential recombinant neuronal actuator. The system was characterized in transcriptional and translational levels in human neuroblastoma SH-SY5Y cells. The mRNA and protein expression levels of nano-ferritin were significant in the activated neurons suggesting that the activity dependent expression patterns of the ferritin also acted as a neuronal cell activation indicator. The system sufficed the need for precise neuronal cell activity specific expression and demonstrated a platform that suggested the use of the nano-ferritin for the study of neuronal cells.
Collapse
Affiliation(s)
- Akara Metasuk
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Phutthamonthon, Nakhon Pathom, 73170, Thailand
| | - Narisorn Kitiyanant
- Stem Cell Research Group, Institute of Molecular Biosciences, Mahidol University, Salaya, Phutthamonthon, Nakhon Pathom, 73170, Thailand
| | - Banthit Chetsawang
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Phutthamonthon, Nakhon Pathom, 73170, Thailand.
| |
Collapse
|
13
|
Sun X, Bernstein MJ, Meng M, Rao S, Sørensen AT, Yao L, Zhang X, Anikeeva PO, Lin Y. Functionally Distinct Neuronal Ensembles within the Memory Engram. Cell 2020; 181:410-423.e17. [PMID: 32187527 PMCID: PMC7166195 DOI: 10.1016/j.cell.2020.02.055] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 10/31/2019] [Accepted: 02/26/2020] [Indexed: 10/24/2022]
Abstract
Memories are believed to be encoded by sparse ensembles of neurons in the brain. However, it remains unclear whether there is functional heterogeneity within individual memory engrams, i.e., if separate neuronal subpopulations encode distinct aspects of the memory and drive memory expression differently. Here, we show that contextual fear memory engrams in the mouse dentate gyrus contain functionally distinct neuronal ensembles, genetically defined by the Fos- or Npas4-dependent transcriptional pathways. The Fos-dependent ensemble promotes memory generalization and receives enhanced excitatory synaptic inputs from the medial entorhinal cortex, which we find itself also mediates generalization. The Npas4-dependent ensemble promotes memory discrimination and receives enhanced inhibitory drive from local cholecystokinin-expressing interneurons, the activity of which is required for discrimination. Our study provides causal evidence for functional heterogeneity within the memory engram and reveals synaptic and circuit mechanisms used by each ensemble to regulate the memory discrimination-generalization balance.
Collapse
Affiliation(s)
- Xiaochen Sun
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Molecular and Cellular Neuroscience Graduate Program, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Max J Bernstein
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Meizhen Meng
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Siyuan Rao
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Andreas T Sørensen
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Li Yao
- State Key Laboratory of Cognitive Neuroscience & Learning and IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Xiaohui Zhang
- State Key Laboratory of Cognitive Neuroscience & Learning and IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China
| | - Polina O Anikeeva
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yingxi Lin
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
14
|
Navabpour S, Kwapis JL, Jarome TJ. A neuroscientist's guide to transgenic mice and other genetic tools. Neurosci Biobehav Rev 2020; 108:732-748. [PMID: 31843544 PMCID: PMC8049509 DOI: 10.1016/j.neubiorev.2019.12.013] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 11/05/2019] [Accepted: 12/09/2019] [Indexed: 12/14/2022]
Abstract
The past decade has produced an explosion in the number and variety of genetic tools available to neuroscientists, resulting in an unprecedented ability to precisely manipulate the genome and epigenome in behaving animals. However, no single resource exists that describes all of the tools available to neuroscientists. Here, we review the genetic, transgenic, and viral techniques that are currently available to probe the complex relationship between genes and cognition. Topics covered include types of traditional transgenic mouse models (knockout, knock-in, reporter lines), inducible systems (Cre-loxP, Tet-On, Tet-Off) and cell- and circuit-specific systems (TetTag, TRAP, DIO-DREADD). Additionally, we provide details on virus-mediated and siRNA/shRNA approaches, as well as a comprehensive discussion of the myriad manipulations that can be made using the CRISPR-Cas9 system, including single base pair editing and spatially- and temporally-regulated gene-specific transcriptional control. Collectively, this review will serve as a guide to assist neuroscientists in identifying and choosing the appropriate genetic tools available to study the complex relationship between the brain and behavior.
Collapse
Affiliation(s)
- Shaghayegh Navabpour
- Fralin Biomedical Research Institute, Translational Biology, Medicine and Health, Roanoke, VA, USA
| | - Janine L Kwapis
- Department of Biology, Pennsylvania State University, College Park, PA, USA; Center for the Molecular Investigation of Neurological Disorders (CMIND), Pennsylvania State University, College Park, PA, USA.
| | - Timothy J Jarome
- Fralin Biomedical Research Institute, Translational Biology, Medicine and Health, Roanoke, VA, USA; Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA; School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.
| |
Collapse
|
15
|
Ebner C, Ledderose J, Zolnik TA, Dominiak SE, Turko P, Papoutsi A, Poirazi P, Eickholt BJ, Vida I, Larkum ME, Sachdev RNS. Optically Induced Calcium-Dependent Gene Activation and Labeling of Active Neurons Using CaMPARI and Cal-Light. Front Synaptic Neurosci 2019; 11:16. [PMID: 31178713 PMCID: PMC6542986 DOI: 10.3389/fnsyn.2019.00016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/03/2019] [Indexed: 02/03/2023] Open
Abstract
The advent of optogenetic methods has made it possible to use endogeneously produced molecules to image and manipulate cellular, subcellular, and synaptic activity. It has also led to the development of photoactivatable calcium-dependent indicators that mark active synapses, neurons, and circuits. Furthermore, calcium-dependent photoactivation can be used to trigger gene expression in active neurons. Here we describe two sets of protocols, one using CaMPARI and a second one using Cal-Light. CaMPARI, a calcium-modulated photoactivatable ratiometric integrator, enables rapid network-wide, tunable, all-optical functional circuit mapping. Cal-Light, a photoactivatable calcium sensor, while slower to respond than CaMPARI, has the capacity to trigger the expression of genes, including effectors, activators, indicators, or other constructs. Here we describe the rationale and provide procedures for using these two calcium-dependent constructs (1) in vitro in dissociated primary neuronal cell cultures (CaMPARI & Cal-Light); (2) in vitro in acute brain slices for circuit mapping (CaMPARI); (3) in vivo for triggering photoconversion or gene expression (CaMPARI & Cal-Light); and finally, (4) for recovering photoconverted neurons post-fixation with immunocytochemistry (CaMPARI). The approaches and protocols we describe are examples of the potential uses of both CaMPARI & Cal-Light. The ability to mark and manipulate neurons that are active during specific epochs of behavior has a vast unexplored experimental potential.
Collapse
Affiliation(s)
- Christian Ebner
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Institute for Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Julia Ledderose
- Institute for Biochemistry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Timothy A Zolnik
- Institute for Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sina E Dominiak
- Institute for Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Paul Turko
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Institute for Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Athanasia Papoutsi
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | - Panayiota Poirazi
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | - Britta J Eickholt
- Institute for Biochemistry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Imre Vida
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Institute for Integrative Neuroanatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Matthew E Larkum
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Institute for Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | | |
Collapse
|
16
|
Discrimination of the hierarchical structure of cortical layers in 2-photon microscopy data by combined unsupervised and supervised machine learning. Sci Rep 2019; 9:7424. [PMID: 31092841 PMCID: PMC6520410 DOI: 10.1038/s41598-019-43432-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 04/18/2019] [Indexed: 12/22/2022] Open
Abstract
The laminar organization of the cerebral cortex is a fundamental characteristic of the brain, with essential implications for cortical function. Due to the rapidly growing amount of high-resolution brain imaging data, a great demand arises for automated and flexible methods for discriminating the laminar texture of the cortex. Here, we propose a combined approach of unsupervised and supervised machine learning to discriminate the hierarchical cortical laminar organization in high-resolution 2-photon microscopic neural image data of mouse brain without observer bias, that is, without the prerequisite of manually labeled training data. For local cortical foci, we modify an unsupervised clustering approach to identify and represent the laminar cortical structure. Subsequently, supervised machine learning is applied to transfer the resulting layer labels across different locations and image data, to ensure the existence of a consistent layer label system. By using neurobiologically meaningful features, the discrimination results are shown to be consistent with the layer classification of the classical Brodmann scheme, and provide additional insight into the structure of the cerebral cortex and its hierarchical organization. Thus, our work paves a new way for studying the anatomical organization of the cerebral cortex, and potentially its functional organization.
Collapse
|
17
|
Activity-dependent visualization and control of neural circuits for courtship behavior in the fly Drosophila melanogaster. Proc Natl Acad Sci U S A 2019; 116:5715-5720. [PMID: 30837311 DOI: 10.1073/pnas.1814628116] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Males of Drosophila melanogaster exhibit stereotypic courtship behavior through which they assess potential mates by processing multimodal sensory information. Although previous studies revealed important neural circuits involved in this process, the full picture of circuits that participate in male courtship remains elusive. Here, we established a genetic tool to visualize or optogenetically reactivate neural circuits activated upon specific behavior, exploiting promoter activity of a neural activity-induced gene Hr38 With this approach, we visualized neural circuits activated in the male brain and the ventral nerve cord when a male interacted with a female. The labeling of neural circuits was additively dependent on inputs from antennae and foreleg tarsi. In addition, neural circuits that express the sex-determining gene fruitless or doublesex were extensively labeled by interaction with a female. Furthermore, optogenetic reactivation of the labeled neural circuits induced courtship posture. With this mapping system, we found that a fruitless-positive neural cluster aSP2 was labeled when a male interacted with a female, in addition to previously characterized neurons. Silencing of neurons including aSP2 led to frequent interruption of courtship and significant reduction of mating success rate without affecting latency to start courtship, suggesting that these neurons are required for courtship persistency important for successful copulation. Overall, these results demonstrate that activity-dependent labeling can be used as a powerful tool not only in vertebrates, but also in invertebrates, to identify neural circuits regulating innate behavior.
Collapse
|
18
|
Schwerdtfeger LA, Tobet SA. From organotypic culture to body-on-a-chip: A neuroendocrine perspective. J Neuroendocrinol 2019; 31:e12650. [PMID: 30307079 DOI: 10.1111/jne.12650] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/02/2018] [Accepted: 10/07/2018] [Indexed: 12/22/2022]
Abstract
The methods used to study neuroendocrinology have been as diverse as the discoveries to come out of the field. Maintaining live neurones outside of a body in vitro was important from the beginning, building on methods that dated back to at least the first decade of the 20th Century. Neurosecretion defines an essential foundation of neuroendocrinology based on work that began in the 1920s and 1930s. Throughout the first half of the 20th Century, many paradigms arose for studying everything from single neurones to whole organs in vitro. Two of these survived as preeminent systems for use throughout the second half of the century: cell cultures and explant systems. Slice cultures and explants that emerged as organotypic technologies included such neuroendocrine organs such as the brain, pituitary, adrenals and intestine. The vast majority of these studies were carried out in static cultures for which media were changed over a time scale of days. Tissues were used for experimental techniques such as electrical recording of neuronal physiology in single cells and observation by live microscopy. When maintained in vitro, many of these systems only partially capture the in vivo physiology of the organ system of interest, often because of a lack of cellular diversity (eg, neuronal cultures lacking glia). Modern microfluidic methodologies show promise for organ systems, ranging from the reproductive to the gastrointestinal to the brain. Moving forward and striving to understand the mechanisms that drive neuroendocrine signalling centrally and peripherally, there will always be a need to consider the heterogeneous cellular compositions of organs in vivo.
Collapse
Affiliation(s)
- Luke A Schwerdtfeger
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Stuart A Tobet
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
- School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado
| |
Collapse
|
19
|
Immediate-Early Promoter-Driven Transgenic Reporter System for Neuroethological Research in a Hemimetabolous Insect. eNeuro 2018; 5:eN-MNT-0061-18. [PMID: 30225346 PMCID: PMC6140108 DOI: 10.1523/eneuro.0061-18.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 07/11/2018] [Accepted: 07/20/2018] [Indexed: 01/04/2023] Open
Abstract
Genes expressed in response to increased neuronal activity are widely used as activity markers in recent behavioral neuroscience. In the present study, we established transgenic reporter system for whole-brain activity mapping in the two-spotted cricket Gryllus bimaculatus, a hemimetabolous insect used in neuroethology and behavioral ecology. In the cricket brain, a homolog of early growth response-1 (Gryllus egr-B) was rapidly induced as an immediate-early gene (IEG) in response to neuronal hyperexcitability. The upstream genomic fragment of Gryllus egr-B contains potential binding sites for transcription factors regulated by various intracellular signaling pathways, as well as core promoter elements conserved across insect/crustacean egr-B homologs. Using the upstream genomic fragment of Gryllus egr-B, we established an IEG promoter-driven transgenic reporter system in the cricket. In the brain of transgenic crickets, the reporter gene (a nuclear-targeted destabilized EYFP) was induced in response to neuronal hyperexcitability. Inducible expression of reporter protein was detected in almost all neurons after neuronal hyperexcitability. Using our novel reporter system, we successfully detected neuronal activation evoked by feeding in the cricket brain. Our IEG promoter-driven activity reporting system allows us to visualize behaviorally relevant neural circuits at cellular resolution in the cricket brain.
Collapse
|
20
|
Abstract
Technologies for measuring the transient Ca2+ spikes that accompany neural signaling have revolutionized our understanding of the brain. Nevertheless, microscopic visualization of Ca2+ spikes on the time scale of neural activity across large brain regions or in thick specimens remains a significant challenge. The recent development of stable integrators of Ca2+, instead of transient reporters, provides an avenue to investigate neural signaling in otherwise challenging systems. Here, we describe an engineered Ca2+-sensing enzyme consisting of a split Tobacco Etch Virus (TEV) protease with each half tethered to a calmodulin or M13 Ca2+ binding domain. This Split TEV, Ca2+ Activated Neuron Recorder (SCANR) remains separate and catalytically incompetent until a spike in cellular Ca2+ triggers its reconstitution and the subsequent turnover of a caged, genetically encoded reporter substrate. We report the identification of a successful Ca2+-sensing split TEV from a library of chimeras and deployment of the enzyme in primary rat hippocampal neurons.
Collapse
Affiliation(s)
- Brianna K. O’Neill
- Department of Chemistry, Stony Brook University, Stony Brook, New York 11794, United States
| | - Scott T. Laughlin
- Department of Chemistry, Stony Brook University, Stony Brook, New York 11794, United States
| |
Collapse
|
21
|
Abstract
Purpose of Review Psychological stress can impact memory systems in several different ways. In individuals with healthy defense and coping systems, stress results in the formation of negatively valenced memories whose ability to induce emotional and somatic distress subsides with time. Vulnerable individuals, however, go on to develop stress-related disorders such as post-traumatic stress disorder (PTSD) and suffer from significant memory abnormalities. Whether expressed as intrusive trauma memories, partial amnesia, or dissociative amnesia, such abnormalities are thought to be the core source of patients' symptoms, which are often debilitating and implicate an entire socio-cognitive-affective spectrum. Recent Findings With this in mind, and focusing on stress-responsive hippocampal microcircuits, this article highlights recent advances in the neurobiology of memory that allow us to (1) isolate and visualize memory circuits, (2) change their activity using genetic tools and state-dependent manipulations, and (3) directly examine their impact on socio-affective circuits and global network connectivity. By integrating these approaches, we are now in a position to address important questions that have troubled psychiatry for a long time-questions such as are traumatic memories special, and why are stress effects on memory diverse. Summary Furthering our fundamental understanding of memory in the framework of adaptive and maladaptive stress responses has the potential to boost the development of new treatments that can benefit patients suffering from psychological trauma.
Collapse
|
22
|
Ben-Shaanan T, Schiller M, Rolls A. Studying brain-regulation of immunity with optogenetics and chemogenetics; A new experimental platform. Brain Behav Immun 2017; 65:1-8. [PMID: 27890661 DOI: 10.1016/j.bbi.2016.11.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 11/22/2016] [Accepted: 11/23/2016] [Indexed: 02/07/2023] Open
Abstract
The interactions between the brain and the immune system are bidirectional. Nevertheless, we have far greater understanding of how the immune system affects the brain than how the brain affects immunity. New technological developments such as optogenetics and chemogenetics (using DREADDs; Designer Receptors Exclusively Activated by Designer Drugs) can bridge this gap in our understanding, as they enable an unprecedented mechanistic and systemic analysis of the communication between the brain and the immune system. In this review, we discuss new experimental approaches for revealing neuronal circuits that can participate in regulation of immunity. In addition, we discuss methods, specifically optogenetics and chemogenetics, that enable targeted neuronal manipulation to reveal how different brain regions affect immunity. We describe how these techniques can be used as an experimental platform to address fundamental questions in psychoneuroimmunology and to understand how neuronal circuits associate with different psychological states can affect physiology.
Collapse
Affiliation(s)
- Tamar Ben-Shaanan
- Department of Immunology, Department of Neuroscience, Rappaport Medical School, Technion - Israel Institute of Technology, Haifa, Israel
| | - Maya Schiller
- Department of Immunology, Department of Neuroscience, Rappaport Medical School, Technion - Israel Institute of Technology, Haifa, Israel
| | - Asya Rolls
- Department of Immunology, Department of Neuroscience, Rappaport Medical School, Technion - Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
23
|
Postnatal Odor Exposure Increases the Strength of Interglomerular Lateral Inhibition onto Olfactory Bulb Tufted Cells. J Neurosci 2017; 36:12321-12327. [PMID: 27927952 DOI: 10.1523/jneurosci.1991-16.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 09/29/2016] [Accepted: 09/30/2016] [Indexed: 11/21/2022] Open
Abstract
Lateral inhibition between pairs of olfactory bulb (OB) mitral cells (MCs) and tufted cells (TCs) is linked to a variety of computations including gain control, decorrelation, and gamma-frequency synchronization. Differential effects of lateral inhibition onto MCs and TCs via distinct lateral inhibitory circuits are one of several recently described circuit-level differences between MCs and TCs that allow each to encode separate olfactory features in parallel. Here, using acute OB slices from mice, we tested whether lateral inhibition is affected by prior odor exposure and if these effects differ between MCs and TCs. We found that early postnatal odor exposure to the M72 glomerulus ligand acetophenone increased the strength of interglomerular lateral inhibition onto TCs, but not MCs, when the M72 glomerulus was stimulated. These increases were specific to exposure to M72 ligands because exposure to hexanal did not increase the strength of M72-mediated lateral inhibition. Therefore, early life experiences may be an important factor shaping TC odor responses. SIGNIFICANCE STATEMENT Responses of olfactory (OB) bulb mitral cells (MCs) and tufted cells (TCs) are known to depend on prior odor exposure, yet the specific circuit mechanisms underlying these experience-dependent changes are unknown. Here, we show that odor exposure alters one particular circuit element, interglomerular lateral inhibition, which is known to be critical for a variety of OB computations. Early postnatal odor exposure to acetophenone, a ligand of M72 olfactory sensory neurons, increases the strength of M72-mediated lateral inhibition onto TCs, but not MCs, that project to nearby glomeruli. These findings add to a growing list of differences between MCs and TCs suggesting that that these two cell types play distinct roles in odor coding.
Collapse
|
24
|
Lee D, Hyun JH, Jung K, Hannan P, Kwon HB. A calcium- and light-gated switch to induce gene expression in activated neurons. Nat Biotechnol 2017. [DOI: 10.1038/nbt.3902] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
25
|
Abstract
Social interactions are often powerful drivers of learning. In female mice, mating creates a long-lasting sensory memory for the pheromones of the stud male that alters neuroendocrine responses to his chemosignals for many weeks. The cellular and synaptic correlates of pheromonal learning, however, remain unclear. We examined local circuit changes in the accessory olfactory bulb (AOB) using targeted ex vivo recordings of mating-activated neurons tagged with a fluorescent reporter. Imprinting led to striking plasticity in the intrinsic membrane excitability of projection neurons (mitral cells, MCs) that dramatically curtailed their responsiveness, suggesting a novel cellular substrate for pheromonal learning. Plasticity was selectively expressed in the MC ensembles activated by the stud male, consistent with formation of memories for specific individuals. Finally, MC excitability gained atypical activity-dependence whose slow dynamics strongly attenuated firing on timescales of several minutes. This unusual form of AOB plasticity may act to filter sustained or repetitive sensory signals. DOI:http://dx.doi.org/10.7554/eLife.25421.001 To navigate social situations, humans and other animals need to remember who they have interacted with and how it went, and adjust their behavior in future encounters accordingly. For example, your physical actions, and even your body’s physiological responses, will be very different when you encounter the last person you kissed instead of the last person you fought with (assuming this is not the same person!). Memories of social interactions can have dramatic consequences. For instance, male mice often kill the offspring of other males. Female mice appear to have adopted a countermeasure to avoid losing a litter of pups to such aggression: they will spontaneously abort a pregnancy when exposed to chemicals called pheromones from unfamiliar males. However, when the female mouse is exposed to the pheromones of the male she mated with she maintains her pregnancy. Exactly how the memories of previous social interactions with the males affect the female’s pheromone responses is not fully understood. To investigate how the female is able to respond differently to different males, Gao et al. recorded the activity of individual neurons taken from the brain tissue of female mice who had recently mated. The recordings showed that previous social experiences produce learning-related changes in the brain of the female mouse that reduce how sensitively pheromone-detecting neurons respond to the chemical cues of the male mate. This suppresses the signals that the neurons would otherwise send to trigger an abortion in response to male pheromones. Gao et al. also used fluorescent tags to identify which neurons in the female’s brain had been activated during mating. This revealed that only those neurons that had been activated by the mate become unresponsive when the cells again encountered his pheromones. This suggests that a set of neurons in the female’s brain records the chemical ‘fingerprint’ of the mate, and can then selectively filter out that mate’s pheromone signals. Many other social interactions, such as parenting, are also strongly shaped by experience. The results presented by Gao et al. may therefore offer wider lessons for understanding how the brain targets different behaviors toward specific individuals. It will also be important to investigate how highly arousing experiences cause such powerful memories to form. This could ultimately help us to better understand – and potentially treat – conditions like post-traumatic stress disorder. DOI:http://dx.doi.org/10.7554/eLife.25421.002
Collapse
Affiliation(s)
- Yuan Gao
- Department of Biology, Boston University, Boston, United States
| | - Carl Budlong
- Department of Biology, Boston University, Boston, United States
| | - Emily Durlacher
- Program in Neuroscience and Behavior, Mount Holyoke College, South Hadley, United States
| | - Ian G Davison
- Department of Biology, Boston University, Boston, United States
| |
Collapse
|
26
|
Comparison of the induction of c-fos-eGFP and Fos protein in the rat spinal cord and hypothalamus resulting from subcutaneous capsaicin or formalin injection. Neuroscience 2017; 356:64-77. [PMID: 28527956 DOI: 10.1016/j.neuroscience.2017.05.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 03/28/2017] [Accepted: 05/09/2017] [Indexed: 12/20/2022]
Abstract
We evaluated whether a c-fos-enhanced green fluorescent protein (eGFP) transgenic rat line, which expresses the c-fos and eGFP fusion gene, can be useful for the study of nociceptive pathways and processing. Capsaicin solution (15%) or formalin (5%) was subcutaneously injected bilaterally into the hind paws (100μL per each paw) of adult male c-fos-eGFP transgenic or wild-type rats. Control rats were injected with ethanol or physiological saline respectively. Transgenic and wild-type rats were perfused at 1.5, 3 and 6h post injection, with some transgenic rats being perfused 24h post injection. A comparison of eGFP in transgenic rats and Fos-like immunoreactivity (LI) in wild-type rats was made in the dorsal spinal cord, paraventricular nucleus (PVN) and supraoptic nucleus (SON). Oxytocin-LI (OXT-LI) was carried out to examine the activation of OXT neurons in the PVN and SON. Following capsaicin or formalin treatment, eGFP was maximally expressed at 6h in the spinal cord and 3h in the PVN and SON, whereas, Fos-LI was maximally expressed at 1.5h in all the regions we analyzed. Induction of eGFP in the OXT neurons was observed after capsaicin or formalin treatment, while Fos-LI in the OXT neurons was observed only after formalin treatment. These results demonstrate that the peak induction of c-fos-eGFP following exposure to acute nociceptive stimuli was delayed by around 1.5-4.5h, but more sensitive than endogenous Fos, suggesting that the c-fos-eGFP rat line can be useful for the study of nociceptive pathways and processing.
Collapse
|
27
|
Temporally precise labeling and control of neuromodulatory circuits in the mammalian brain. Nat Methods 2017; 14:495-503. [PMID: 28369042 DOI: 10.1038/nmeth.4234] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 02/18/2017] [Indexed: 11/08/2022]
Abstract
Few tools exist to visualize and manipulate neurons that are targets of neuromodulators. We present iTango, a light- and ligand-gated gene expression system based on a light-inducible split tobacco etch virus protease. Cells expressing the iTango system exhibit increased expression of a marker gene in the presence of dopamine and blue-light exposure, both in vitro and in vivo. We demonstrated the iTango system in a behaviorally relevant context, by inducing expression of optogenetic tools in neurons under dopaminergic control during a behavior of interest. We thereby gained optogenetic control of these behaviorally relevant neurons. We applied the iTango system to decipher the roles of two classes of dopaminergic neurons in the mouse nucleus accumbens in a sensitized locomotor response to cocaine. Thus, the iTango platform allows for control of neuromodulatory circuits in a genetically and functionally defined manner with spatial and temporal precision.
Collapse
|
28
|
Cho JH, Rendall SD, Gray JM. Brain-wide maps of Fos expression during fear learning and recall. ACTA ACUST UNITED AC 2017; 24:169-181. [PMID: 28331016 PMCID: PMC5362696 DOI: 10.1101/lm.044446.116] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 02/23/2017] [Indexed: 12/12/2022]
Abstract
Fos induction during learning labels neuronal ensembles in the hippocampus that encode a specific physical environment, revealing a memory trace. In the cortex and other regions, the extent to which Fos induction during learning reveals specific sensory representations is unknown. Here we generate high-quality brain-wide maps of Fos mRNA expression during auditory fear conditioning and recall in the setting of the home cage. These maps reveal a brain-wide pattern of Fos induction that is remarkably similar among fear conditioning, shock-only, tone-only, and fear recall conditions, casting doubt on the idea that Fos reveals auditory-specific sensory representations. Indeed, novel auditory tones lead to as much gene induction in visual as in auditory cortex, while familiar (nonconditioned) tones do not appreciably induce Fos anywhere in the brain. Fos expression levels do not correlate with physical activity, suggesting that they are not determined by behavioral activity-driven alterations in sensory experience. In the thalamus, Fos is induced more prominently in limbic than in sensory relay nuclei, suggesting that Fos may be most sensitive to emotional state. Thus, our data suggest that Fos expression during simple associative learning labels ensembles activated generally by arousal rather than specifically by a particular sensory cue.
Collapse
Affiliation(s)
- Jin-Hyung Cho
- Genetics Department, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Sam D Rendall
- Genetics Department, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Jesse M Gray
- Genetics Department, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
29
|
Mastwal S, Cao V, Wang KH. Genetic Feedback Regulation of Frontal Cortical Neuronal Ensembles Through Activity-Dependent Arc Expression and Dopaminergic Input. Front Neural Circuits 2016; 10:100. [PMID: 27999532 PMCID: PMC5138219 DOI: 10.3389/fncir.2016.00100] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/23/2016] [Indexed: 12/15/2022] Open
Abstract
Mental functions involve coordinated activities of specific neuronal ensembles that are embedded in complex brain circuits. Aberrant neuronal ensemble dynamics is thought to form the neurobiological basis of mental disorders. A major challenge in mental health research is to identify these cellular ensembles and determine what molecular mechanisms constrain their emergence and consolidation during development and learning. Here, we provide a perspective based on recent studies that use activity-dependent gene Arc/Arg3.1 as a cellular marker to identify neuronal ensembles and a molecular probe to modulate circuit functions. These studies have demonstrated that the transcription of Arc is activated in selective groups of frontal cortical neurons in response to specific behavioral tasks. Arc expression regulates the persistent firing of individual neurons and predicts the consolidation of neuronal ensembles during repeated learning. Therefore, the Arc pathway represents a prototypical example of activity-dependent genetic feedback regulation of neuronal ensembles. The activation of this pathway in the frontal cortex starts during early postnatal development and requires dopaminergic (DA) input. Conversely, genetic disruption of Arc leads to a hypoactive mesofrontal dopamine circuit and its related cognitive deficit. This mutual interaction suggests an auto-regulatory mechanism to amplify the impact of neuromodulators and activity-regulated genes during postnatal development. Such a mechanism may contribute to the association of mutations in dopamine and Arc pathways with neurodevelopmental psychiatric disorders. As the mesofrontal dopamine circuit shows extensive activity-dependent developmental plasticity, activity-guided modulation of DA projections or Arc ensembles during development may help to repair circuit deficits related to neuropsychiatric disorders.
Collapse
Affiliation(s)
- Surjeet Mastwal
- Unit on Neural Circuits and Adaptive Behaviors, Clinical and Translational Neuroscience Branch, National Institute of Mental Health Bethesda, MD, USA
| | - Vania Cao
- Unit on Neural Circuits and Adaptive Behaviors, Clinical and Translational Neuroscience Branch, National Institute of Mental Health Bethesda, MD, USA
| | - Kuan Hong Wang
- Unit on Neural Circuits and Adaptive Behaviors, Clinical and Translational Neuroscience Branch, National Institute of Mental Health Bethesda, MD, USA
| |
Collapse
|
30
|
Abstract
UNLABELLED Comprehensive analysis of brain function depends on understanding the dynamics of diverse neural signaling processes over large tissue volumes in intact animals and humans. Most existing approaches to measuring brain signaling suffer from limited tissue penetration, poor resolution, or lack of specificity for well-defined neural events. Here we discuss a new brain activity mapping method that overcomes some of these problems by combining MRI with contrast agents sensitive to neural signaling. The goal of this "molecular fMRI" approach is to permit noninvasive whole-brain neuroimaging with specificity and resolution approaching current optical neuroimaging methods. In this article, we describe the context and need for molecular fMRI as well as the state of the technology today. We explain how major types of MRI probes work and how they can be sensitized to neurobiological processes, such as neurotransmitter release, calcium signaling, and gene expression changes. We comment both on past work in the field and on challenges and promising avenues for future development. SIGNIFICANCE STATEMENT Brain researchers currently have a choice between measuring neural activity using cellular-level recording techniques, such as electrophysiology and optical imaging, or whole-brain imaging methods, such as fMRI. Cellular level methods are precise but only address a small portion of mammalian brains; on the other hand, whole-brain neuroimaging techniques provide very little specificity for neural pathways or signaling components of interest. The molecular fMRI techniques we discuss have particular potential to combine the specificity of cellular-level measurements with the noninvasive whole-brain coverage of fMRI. On the other hand, molecular fMRI is only just getting off the ground. This article aims to offer a snapshot of the status and future prospects for development of molecular fMRI techniques.
Collapse
|
31
|
Abstract
UNLABELLED The proper growth and arborization of dendrites in response to sensory experience are essential for neural connectivity and information processing in the brain. Although neuronal activity is important for sculpting dendrite morphology, the underlying molecular mechanisms are not well understood. Here, we report that cyclin-dependent kinase 5 (Cdk5)-mediated transcriptional regulation is a key mechanism that controls activity-dependent dendrite development in cultured rat neurons. During membrane depolarization, Cdk5 accumulates in the nucleus to regulate the expression of a subset of genes, including that of the neurotrophin brain-derived neurotrophic factor, for subsequent dendritic growth. Furthermore, Cdk5 function is mediated through the phosphorylation of methyl-CpG-binding protein 2, a key transcriptional repressor that is mutated in the mental disorder Rett syndrome. These findings collectively suggest that the nuclear import of Cdk5 is crucial for activity-dependent dendrite development by regulating neuronal gene transcription during neural development. SIGNIFICANCE STATEMENT Neural activity directs dendrite development through the regulation of gene transcription. However, how molecular signals link extracellular stimuli to the transcriptional program in the nucleus remains unclear. Here, we demonstrate that neuronal activity stimulates the translocation of the kinase Cdk5 from the cytoplasmic compartment into the nucleus; furthermore, the nuclear localization of Cdk5 is required for dendrite development in cultured neurons. Genome-wide transcriptome analysis shows that Cdk5 deficiency specifically disrupts activity-dependent gene transcription of bdnf. The action of Cdk5 is mediated through the modulation of the transcriptional repressor methyl-CpG-binding protein 2. Therefore, this study elucidates the role of nuclear Cdk5 in the regulation of activity-dependent gene transcription and dendritic growth.
Collapse
|
32
|
Macpherson LJ, Zaharieva EE, Kearney PJ, Alpert MH, Lin TY, Turan Z, Lee CH, Gallio M. Dynamic labelling of neural connections in multiple colours by trans-synaptic fluorescence complementation. Nat Commun 2015; 6:10024. [PMID: 26635273 PMCID: PMC4686661 DOI: 10.1038/ncomms10024] [Citation(s) in RCA: 148] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 10/27/2015] [Indexed: 12/19/2022] Open
Abstract
Determining the pattern of activity of individual connections within a neural circuit could provide insights into the computational processes that underlie brain function. Here, we develop new strategies to label active synapses by trans-synaptic fluorescence complementation in Drosophila. First, we demonstrate that a synaptobrevin-GRASP chimera functions as a powerful activity-dependent marker for synapses in vivo. Next, we create cyan and yellow variants, achieving activity-dependent, multi-colour fluorescence reconstitution across synapses (X-RASP). Our system allows for the first time retrospective labelling of synapses (rather than whole neurons) based on their activity, in multiple colours, in the same animal. As individual synapses often act as computational units in the brain, our method will promote the design of experiments that are not possible using existing techniques. Moreover, our strategies are easily adaptable to circuit mapping in any genetic system.
Collapse
Affiliation(s)
- Lindsey J Macpherson
- Howard Hughes Medical Institute and Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York 10032, USA
| | - Emanuela E Zaharieva
- Department of Neurobiology, Northwestern University, Evanston, Illinois 60208, USA
| | - Patrick J Kearney
- Department of Neurobiology, Northwestern University, Evanston, Illinois 60208, USA
| | - Michael H Alpert
- Department of Neurobiology, Northwestern University, Evanston, Illinois 60208, USA
| | - Tzu-Yang Lin
- Section on Neuronal Connectivity, Laboratory of Gene Regulation and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Zeynep Turan
- Howard Hughes Medical Institute and Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York 10032, USA
| | - Chi-Hon Lee
- Section on Neuronal Connectivity, Laboratory of Gene Regulation and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Marco Gallio
- Department of Neurobiology, Northwestern University, Evanston, Illinois 60208, USA
| |
Collapse
|
33
|
Wagener RJ, Witte M, Guy J, Mingo-Moreno N, Kügler S, Staiger JF. Thalamocortical Connections Drive Intracortical Activation of Functional Columns in the Mislaminated Reeler Somatosensory Cortex. Cereb Cortex 2015; 26:820-37. [PMID: 26564256 PMCID: PMC4712806 DOI: 10.1093/cercor/bhv257] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Neuronal wiring is key to proper neural information processing. Tactile information from the rodent's whiskers reaches the cortex via distinct anatomical pathways. The lemniscal pathway relays whisking and touch information from the ventral posteromedial thalamic nucleus to layer IV of the primary somatosensory “barrel” cortex. The disorganized neocortex of the reeler mouse is a model system that should severely compromise the ingrowth of thalamocortical axons (TCAs) into the cortex. Moreover, it could disrupt intracortical wiring. We found that neuronal intermingling within the reeler barrel cortex substantially exceeded previous descriptions, leading to the loss of layers. However, viral tracing revealed that TCAs still specifically targeted transgenically labeled spiny layer IV neurons. Slice electrophysiology and optogenetics proved that these connections represent functional synapses. In addition, we assessed intracortical activation via immediate-early-gene expression resulting from a behavioral exploration task. The cellular composition of activated neuronal ensembles suggests extensive similarities in intracolumnar information processing in the wild-type and reeler brains. We conclude that extensive ectopic positioning of neuronal partners can be compensated for by cell-autonomous mechanisms that allow for the establishment of proper connectivity. Thus, genetic neuronal fate seems to be of greater importance for correct cortical wiring than radial neuronal position.
Collapse
Affiliation(s)
- Robin J Wagener
- Institute for Neuroanatomy, Universitätsmedizin Göttingen, Georg-August-Universität Göttingen, 37075 Göttingen, Germany
| | - Mirko Witte
- Institute for Neuroanatomy, Universitätsmedizin Göttingen, Georg-August-Universität Göttingen, 37075 Göttingen, Germany
| | - Julien Guy
- Institute for Neuroanatomy, Universitätsmedizin Göttingen, Georg-August-Universität Göttingen, 37075 Göttingen, Germany
| | - Nieves Mingo-Moreno
- Institute for Neuroanatomy, Universitätsmedizin Göttingen, Georg-August-Universität Göttingen, 37075 Göttingen, Germany Center Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Sebastian Kügler
- Center Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany Department of Neurology, Universitätsmedizin Göttingen, Georg-August-Universität Göttingen, 37075 Göttingen, Germany
| | - Jochen F Staiger
- Institute for Neuroanatomy, Universitätsmedizin Göttingen, Georg-August-Universität Göttingen, 37075 Göttingen, Germany Center Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| |
Collapse
|
34
|
Morland RH, Novejarque A, Huang W, Wodarski R, Denk F, Dawes JD, Pheby T, McMahon SB, Rice AS. Short-term effect of acute and repeated urinary bladder inflammation on thigmotactic behaviour in the laboratory rat. F1000Res 2015; 4:109. [PMID: 27158443 PMCID: PMC4850861 DOI: 10.12688/f1000research.6255.1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/28/2015] [Indexed: 12/13/2022] Open
Abstract
Understanding the non-sensory components of the pain experience is crucial to developing effective treatments for pain conditions. Chronic pain is associated with increased incidence of anxio-depressive disorders, and patients often report feelings of vulnerability which can decrease quality of life. In animal models of pain, observation of behaviours such as thigmotaxis can be used to detect such affective disturbances by exploiting the influence of nociceptive stimuli on the innate behavioural conflict between exploration of a novel space and predator avoidance behaviour. This study investigates whether acute and repeated bladder inflammation in adult female Wistar rats increases thigmotactic behaviour in the open field paradigm, and aims to determine whether this correlates with activation in the central amygdala, as measured by c-Fos immunoreactivity. Additionally, up-regulation of inflammatory mediators in the urinary bladder was measured using RT-qPCR array featuring 92 transcripts to examine how local mediators change under experimental conditions. We found acute but not repeated turpentine inflammation of the bladder increased thigmotactic behaviour (decreased frequency of entry to the inner zone) in the open field paradigm, a result that was also observed in the catheter-only instrumentation group. Decreases in locomotor activity were also observed in both models in turpentine and instrumentation groups. No differences were observed in c-Fos activation, although a general increased in activation along the rostro-caudal axis was seen. Inflammatory mediator up-regulation was greatest following acute inflammation, with CCL12, CCL7, and IL-1β significantly up-regulated in both conditions when compared to naïve tissue. These results suggest that acute catheterisation, with or without turpentine inflammation, induces affective alterations detectable in the open field paradigm accompanied by up-regulation of multiple inflammatory mediators.
Collapse
Affiliation(s)
- Rosemary H Morland
- Pain Research Group, Department of Surgery and Cancer, Faculty of Medicine, Imperial College , London, UK
| | - Amparo Novejarque
- Pain Research Group, Department of Surgery and Cancer, Faculty of Medicine, Imperial College , London, UK
| | - Wenlong Huang
- Pain Research Group, Department of Surgery and Cancer, Faculty of Medicine, Imperial College , London, UK
| | - Rachel Wodarski
- Pain Research Group, Department of Surgery and Cancer, Faculty of Medicine, Imperial College , London, UK
| | - Franziska Denk
- Wolfson Centre for Age Related Disease, King's College London, London, UK
| | - John D Dawes
- The Nuffield Department of Clinical Neurosciences, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Tim Pheby
- Pain Research Group, Department of Surgery and Cancer, Faculty of Medicine, Imperial College , London, UK
| | - Stephen B McMahon
- Wolfson Centre for Age Related Disease, King's College London, London, UK
| | - Andrew Sc Rice
- Pain Research Group, Department of Surgery and Cancer, Faculty of Medicine, Imperial College , London, UK
| |
Collapse
|
35
|
Ichijo H, Hamada M, Takahashi S, Kobayashi M, Nagai T, Toyama T, Kawaguchi M. Lateralization, maturation, and anteroposterior topography in the lateral habenula revealed by ZIF268/EGR1 immunoreactivity and labeling history of neuronal activity. Neurosci Res 2015; 95:27-37. [PMID: 25637311 DOI: 10.1016/j.neures.2015.01.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 01/16/2015] [Accepted: 01/16/2015] [Indexed: 11/28/2022]
Abstract
We report habenular lateralization in a simple transgenic mouse model used for labeling a facet of neuronal activity history. A transgenic construct comprised of a zif268/egr1 immediate-early gene promoter and a gene for normal Venus fluorescent protein with a membrane tag converted promoter activity into long-life fluorescent proteins, which was thought to describe a facet of neuronal activity history by summing neuronal activity. In addition to mapping the immediate-early gene-immunopositive cells, this method helped demonstrate the functionality of the lateral habenular nucleus (LHb). During postnatal development, the LHb was activated between postnatal days 10 and 16. The water-immersion restraint stress also activated the LHb over a similar period. LHb activation was functionally lateralized, but had no directional bias at the population level. Moreover, the posterior LHb was activated in the early stage after the stress, while the anterior LHb was activated in the later stage. Our results indicate lateralization, maturation, and anteroposterior topography of the LHb during postnatal development and the stress response.
Collapse
Affiliation(s)
- Hiroyuki Ichijo
- Department of Anatomy, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan; Department of Anatomy and Embryology, Division of Biomedical Science, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan.
| | - Michito Hamada
- Department of Anatomy and Embryology, Division of Biomedical Science, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan
| | - Satoru Takahashi
- Department of Anatomy and Embryology, Division of Biomedical Science, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan; Laboratory Animal Resource Center, Division of Biomedical Science, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan
| | - Makoto Kobayashi
- Department of Molecular and Developmental Biology, Division of Biomedical Science, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan
| | - Takeharu Nagai
- The Institute of Scientific and Industrial Research, Osaka University, Ibaraki 567-0047, Japan
| | - Tomoko Toyama
- Department of Anatomy and Embryology, Division of Biomedical Science, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan
| | - Masahumi Kawaguchi
- Department of Anatomy, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| |
Collapse
|
36
|
Inoue M, Takeuchi A, Horigane SI, Ohkura M, Gengyo-Ando K, Fujii H, Kamijo S, Takemoto-Kimura S, Kano M, Nakai J, Kitamura K, Bito H. Rational design of a high-affinity, fast, red calcium indicator R-CaMP2. Nat Methods 2014; 12:64-70. [PMID: 25419959 DOI: 10.1038/nmeth.3185] [Citation(s) in RCA: 202] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 10/25/2014] [Indexed: 12/11/2022]
Abstract
Fluorescent Ca(2+) reporters are widely used as readouts of neuronal activities. Here we designed R-CaMP2, a high-affinity red genetically encoded calcium indicator (GECI) with a Hill coefficient near 1. Use of the calmodulin-binding sequence of CaMKK-α and CaMKK-β in lieu of an M13 sequence resulted in threefold faster rise and decay times of Ca(2+) transients than R-CaMP1.07. These features allowed resolving single action potentials (APs) and recording fast AP trains up to 20-40 Hz in cortical slices. Somatic and synaptic activities of a cortical neuronal ensemble in vivo were imaged with similar efficacy as with previously reported sensitive green GECIs. Combining green and red GECIs, we successfully achieved dual-color monitoring of neuronal activities of distinct cell types, both in the mouse cortex and in freely moving Caenorhabditis elegans. Dual imaging using R-CaMP2 and green GECIs provides a powerful means to interrogate orthogonal and hierarchical neuronal ensembles in vivo.
Collapse
Affiliation(s)
- Masatoshi Inoue
- 1] Department of Neurochemistry, Graduate School of Medicine, University of Tokyo, Tokyo, Japan. [2] Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo, Japan
| | - Atsuya Takeuchi
- Department of Neurophysiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Shin-ichiro Horigane
- Department of Neurochemistry, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | | | | | - Hajime Fujii
- Department of Neurochemistry, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Satoshi Kamijo
- 1] Department of Neurochemistry, Graduate School of Medicine, University of Tokyo, Tokyo, Japan. [2] Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo, Japan
| | - Sayaka Takemoto-Kimura
- 1] Department of Neurochemistry, Graduate School of Medicine, University of Tokyo, Tokyo, Japan. [2] Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency, Saitama, Japan
| | - Masanobu Kano
- Department of Neurophysiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Junichi Nakai
- Brain Science Institute, Saitama University, Saitama, Japan
| | - Kazuo Kitamura
- 1] Department of Neurophysiology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan. [2] Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science and Technology Agency, Saitama, Japan
| | - Haruhiko Bito
- 1] Department of Neurochemistry, Graduate School of Medicine, University of Tokyo, Tokyo, Japan. [2] Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo, Japan
| |
Collapse
|
37
|
Ghitani N, Bayguinov PO, Ma Y, Jackson MB. Single-trial imaging of spikes and synaptic potentials in single neurons in brain slices with genetically encoded hybrid voltage sensor. J Neurophysiol 2014; 113:1249-59. [PMID: 25411462 DOI: 10.1152/jn.00691.2014] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Genetically encoded voltage sensors expand the optogenetics toolkit into the important realm of electrical recording, enabling researchers to study the dynamic activity of complex neural circuits in real time. However, these probes have thus far performed poorly when tested in intact neural circuits. Hybrid voltage sensors (hVOS) enable the imaging of voltage by harnessing the resonant energy transfer that occurs between a genetically encoded component, a membrane-tethered fluorescent protein that serves as a donor, and a small charged molecule, dipicrylamine, which serves as an acceptor. hVOS generates optical signals as a result of voltage-induced changes in donor-acceptor distance. We expressed the hVOS probe in mouse brain by in utero electroporation and in transgenic mice with a neuronal promoter. Under conditions favoring sparse labeling we could visualize single-labeled neurons. hVOS imaging reported electrically evoked fluorescence changes from individual neurons in slices from entorhinal cortex, somatosensory cortex, and hippocampus. These fluorescence signals tracked action potentials in individual neurons in a single trial with excellent temporal fidelity, producing changes that exceeded background noise by as much as 16-fold. Subthreshold synaptic potentials were detected in single trials in multiple distinct cells simultaneously. We followed signal propagation between different cells within one field of view and between dendrites and somata of the same cell. hVOS imaging thus provides a tool for high-resolution recording of electrical activity from genetically targeted cells in intact neuronal circuits.
Collapse
Affiliation(s)
- Nima Ghitani
- Neuroscience Training Program, University of Wisconsin, Madison, Wisconsin
| | - Peter O Bayguinov
- Department of Neuroscience, University of Wisconsin, Madison, Wisconsin
| | - Yihe Ma
- Physiology Graduate Training Program, University of Wisconsin, Madison, Wisconsin; and
| | - Meyer B Jackson
- Neuroscience Training Program, University of Wisconsin, Madison, Wisconsin; Physiology Graduate Training Program, University of Wisconsin, Madison, Wisconsin; and Department of Neuroscience, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
38
|
Cruz FC, Javier Rubio F, Hope BT. Using c-fos to study neuronal ensembles in corticostriatal circuitry of addiction. Brain Res 2014; 1628:157-73. [PMID: 25446457 DOI: 10.1016/j.brainres.2014.11.005] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 10/27/2014] [Accepted: 11/01/2014] [Indexed: 01/02/2023]
Abstract
Learned associations between drugs and environment play an important role in addiction and are thought to be encoded within specific patterns of sparsely distributed neurons called neuronal ensembles. This hypothesis is supported by correlational data from in vivo electrophysiology and cellular imaging studies in relapse models in rodents. In particular, cellular imaging with the immediate early gene c-fos and its protein product Fos has been used to identify sparsely distributed neurons that were strongly activated during conditioned drug behaviors such as drug self-administration and context- and cue-induced reinstatement of drug seeking. Here we review how Fos and the c-fos promoter have been employed to demonstrate causal roles for Fos-expressing neuronal ensembles in prefrontal cortex and nucleus accumbens in conditioned drug behaviors. This work has allowed identification of unique molecular and electrophysiological alterations within Fos-expressing neuronal ensembles that may contribute to the development and expression of learned associations in addiction.
Collapse
Affiliation(s)
- Fabio C Cruz
- Behavioral Neuroscience Branch, IRP/NIDA/NIH/DHHS, 251 Bayview Blvd, Suite 200, Baltimore, MD 21224, United States
| | - F Javier Rubio
- Behavioral Neuroscience Branch, IRP/NIDA/NIH/DHHS, 251 Bayview Blvd, Suite 200, Baltimore, MD 21224, United States
| | - Bruce T Hope
- Behavioral Neuroscience Branch, IRP/NIDA/NIH/DHHS, 251 Bayview Blvd, Suite 200, Baltimore, MD 21224, United States.
| |
Collapse
|
39
|
Wouterlood FG, Bloem B, Mansvelder HD, Luchicchi A, Deisseroth K. A fourth generation of neuroanatomical tracing techniques: exploiting the offspring of genetic engineering. J Neurosci Methods 2014; 235:331-48. [PMID: 25107853 DOI: 10.1016/j.jneumeth.2014.07.021] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 07/28/2014] [Accepted: 07/29/2014] [Indexed: 11/18/2022]
Abstract
The first three generations of neuroanatomical tract-tracing methods include, respectively, techniques exploiting degeneration, retrograde cellular transport and anterograde cellular transport. This paper reviews the most recent development in third-generation tracing, i.e., neurochemical fingerprinting based on BDA tracing, and continues with an emerging tracing technique called here 'selective fluorescent protein expression' that in our view belongs to an entirely new 'fourth-generation' class. Tracing techniques in this class lean on gene expression technology designed to 'label' projections exclusively originating from neurons expressing a very specific molecular phenotype. Genetically engineered mice that express cre-recombinase in a neurochemically specific neuronal population receive into a brain locus of interest an injection of an adeno-associated virus (AAV) carrying a double-floxed promoter-eYFP DNA sequence. After transfection this sequence is expressed only in neurons metabolizing recombinase protein. These particular neurons promptly start manufacturing the fluorescent protein which then accumulates and labels to full detail all the neuronal processes, including fibers and terminal arborizations. All other neurons remain optically 'dark'. The AAV is not replicated by the neurons, prohibiting intracerebral spread of 'infection'. The essence is that the fiber projections of discrete subpopulations of neurochemically specific neurons can be traced in full detail. One condition is that the transgenic mouse strain is recombinase-perfect. We illustrate selective fluorescent protein expression in parvalbumin-cre (PV-cre) mice and choline acetyltransferase-cre (ChAT-cre) mice. In addition we compare this novel tracing technique with observations in brains of native PV mice and ChAT-GFP mice. We include a note on tracing techniques using viruses.
Collapse
Affiliation(s)
- Floris G Wouterlood
- Department of Anatomy and Neurosciences, Neuroscience Campus Amsterdam, Vrije University Medical Center, Amsterdam, The Netherlands.
| | - Bernard Bloem
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands; Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Huibert D Mansvelder
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Antonio Luchicchi
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research (CNCR), Neuroscience Campus Amsterdam, VU University, Amsterdam, The Netherlands
| | - Karl Deisseroth
- Bioengineering Department, James E. Clark Center, Stanford University, Stanford, CA, USA
| |
Collapse
|
40
|
Synaptic modifications in the medial prefrontal cortex in susceptibility and resilience to stress. J Neurosci 2014; 34:7485-92. [PMID: 24872553 DOI: 10.1523/jneurosci.5294-13.2014] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
When facing stress, most individuals are resilient whereas others are prone to developing mood disorders. The brain mechanisms underlying such divergent behavioral responses remain unclear. Here we used the learned helplessness procedure in mice to examine the role of the medial prefrontal cortex (mPFC), a brain region highly implicated in both clinical and animal models of depression, in adaptive and maladaptive behavioral responses to stress. We found that uncontrollable and inescapable stress induced behavioral state-dependent changes in the excitatory synapses onto a subset of mPFC neurons: those that were activated during behavioral responses as indicated by their expression of the activity reporter c-Fos. Whereas synaptic potentiation was linked to learned helplessness, a depression-like behavior, synaptic weakening, was associated with resilience to stress. Notably, enhancing the activity of mPFC neurons using a chemical-genetic method was sufficient to convert the resilient behavior into helplessness. Our results provide direct evidence that mPFC dysfunction is linked to maladaptive behavioral responses to stress, and suggest that enhanced excitatory synaptic drive onto mPFC neurons may underlie the previously reported hyperactivity of this brain region in depression.
Collapse
|
41
|
Abstract
The brain encodes information about past experience in specific populations of neurons that communicate with one another by firing action potentials. Studies of experience-dependent neural plasticity have largely focused on individual synaptic changes in response to neuronal input. Indicative of the neuronal output transmitted to downstream neurons, persistent firing patterns are affected by prior experience in selective neuronal populations. However, little is known about the molecular and cellular mechanisms by which experience-related persistent firing patterns are regulated in specific neuronal populations. Using frontal cortical slices prepared from transgenic mice carrying a fluorescent reporter of Arc gene expression, this study investigates how behavioral experience and the activity-regulated Arc gene affect patterns of neuronal firing. We found that motor training increases Arc expression in subsets of excitatory neurons. Those neurons exhibit persistent firing in contrast to Arc-negative neurons from the same mice or neurons from the untrained mice. Furthermore, in mice carrying genetic deletion of Arc, the frontal cortical circuitry is still in place to initiate experience-dependent gene expression, but the level of persistent firing thereafter is diminished. Finally, our results showed that the emergence of persistent activity is associated with Arc-dependent changes in the function of NMDA-type glutamate receptors, rather than changes in AMPA-type receptors or membrane excitability. Our findings therefore reveal an Arc-dependent molecular pathway by which gene-experience interaction regulates the emergence of persistent firing patterns in specific neuronal populations.
Collapse
|
42
|
Encoding and storage of spatial information in the retrosplenial cortex. Proc Natl Acad Sci U S A 2014; 111:8661-6. [PMID: 24912150 DOI: 10.1073/pnas.1313222111] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The retrosplenial cortex (RSC) is part of a network of interconnected cortical, hippocampal, and thalamic structures harboring spatially modulated neurons. The RSC contains head direction cells and connects to the parahippocampal region and anterior thalamus. Manipulations of the RSC can affect spatial and contextual tasks. A considerable amount of evidence implicates the role of the RSC in spatial navigation, but it is unclear whether this structure actually encodes or stores spatial information. We used a transgenic mouse in which the expression of green fluorescent protein was under the control of the immediate early gene c-fos promoter as well as time-lapse two-photon in vivo imaging to monitor neuronal activation triggered by spatial learning in the Morris water maze. We uncovered a repetitive pattern of cell activation in the RSC consistent with the hypothesis that during spatial learning an experience-dependent memory trace is formed in this structure. In support of this hypothesis, we also report three other observations. First, temporary RSC inactivation disrupts performance in a spatial learning task. Second, we show that overexpressing the transcription factor CREB in the RSC with a viral vector, a manipulation known to enhance memory consolidation in other circuits, results in spatial memory enhancements. Third, silencing the viral CREB-expressing neurons with the allatostatin system occludes the spatial memory enhancement. Taken together, these results indicate that the retrosplenial cortex engages in the formation and storage of memory traces for spatial information.
Collapse
|
43
|
Katoh A, Shoguchi K, Matsuoka H, Yoshimura M, Ohkubo JI, Matsuura T, Maruyama T, Ishikura T, Aritomi T, Fujihara H, Hashimoto H, Suzuki H, Murphy D, Ueta Y. Fluorescent visualisation of the hypothalamic oxytocin neurones activated by cholecystokinin-8 in rats expressing c-fos-enhanced green fluorescent protein and oxytocin-monomeric red fluorescent protein 1 fusion transgenes. J Neuroendocrinol 2014; 26:341-7. [PMID: 24730419 DOI: 10.1111/jne.12150] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2013] [Revised: 02/27/2014] [Accepted: 03/15/2014] [Indexed: 12/01/2022]
Abstract
The up-regulation of c-fos gene expression is widely used as a marker of neuronal activation elicited by various stimuli. Anatomically precise observation of c-fos gene products can be achieved at the RNA level by in situ hybridisation or at the protein level by immunocytochemistry. Both of these methods are time and labour intensive. We have developed a novel transgenic rat system that enables the trivial visualisation of c-fos expression using an enhanced green fluorescent protein (eGFP) tag. These rats express a transgene consisting of c-fos gene regulatory sequences that drive the expression of a c-fos-eGFP fusion protein. In c-fos-eGFP transgenic rats, robust nuclear eGFP fluorescence was observed in osmosensitive brain regions 90 min after i.p. administration of hypertonic saline. Nuclear eGFP fluorescence was also observed in the supraoptic nucleus (SON) and paraventricular nucleus (PVN) 90 min after i.p. administration of cholecystokinin (CCK)-8, which selectively activates oxytocin (OXT)-secreting neurones in the hypothalamus. In double transgenic rats that express c-fos-eGFP and an OXT-monomeric red fluorescent protein 1 (mRFP1) fusion gene, almost all mRFP1-positive neurones in the SON and PVN expressed nuclear eGFP fluorescence 90 min after i.p. administration of CCK-8. It is possible that not only a plane image, but also three-dimensional reconstruction image may identify cytoplasmic vesicles in an activated neurone at the same time.
Collapse
Affiliation(s)
- A Katoh
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan; Otorhynolaryngology-Head and Neck Surgery, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan; Department of Otorhynolaryngology, National Kyushu Medical Center, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Vousden DA, Epp J, Okuno H, Nieman BJ, van Eede M, Dazai J, Ragan T, Bito H, Frankland PW, Lerch JP, Henkelman RM. Whole-brain mapping of behaviourally induced neural activation in mice. Brain Struct Funct 2014; 220:2043-57. [PMID: 24760545 DOI: 10.1007/s00429-014-0774-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 04/03/2014] [Indexed: 10/25/2022]
Abstract
The ability to visualize behaviourally evoked neural activity patterns across the rodent brain is essential for understanding the distributed brain networks mediating particular behaviours. However, current imaging methods are limited in their spatial resolution and/or ability to obtain brain-wide coverage of functional activity. Here, we describe a new automated method for obtaining cellular-level, whole-brain maps of behaviourally induced neural activity in the mouse. This method combines the use of transgenic immediate-early gene reporter mice to visualize neural activity; serial two-photon tomography to image the entire brain at cellular resolution; advanced image processing algorithms to count the activated neurons and align the datasets to the Allen Mouse Brain Atlas; and statistical analysis to identify the network of activated brain regions evoked by behaviour. We demonstrate the use of this approach to determine the whole-brain networks activated during the retrieval of fear memories. Consistent with previous studies, we identified a large network of amygdalar, hippocampal, and neocortical brain regions implicated in fear memory retrieval. Our proposed methods can thus be used to map cellular networks involved in the expression of normal behaviours as well as to investigate in depth circuit dysfunction in mouse models of neurobiological disease.
Collapse
Affiliation(s)
- Dulcie A Vousden
- Mouse Imaging Centre, The Hospital for Sick Children, 25 Orde St., Toronto, ON M5T 3H7, Canada,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Kawashima T, Okuno H, Bito H. A new era for functional labeling of neurons: activity-dependent promoters have come of age. Front Neural Circuits 2014; 8:37. [PMID: 24795570 PMCID: PMC4005930 DOI: 10.3389/fncir.2014.00037] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Accepted: 04/01/2014] [Indexed: 12/03/2022] Open
Abstract
Genetic labeling of neurons with a specific response feature is an emerging technology for precise dissection of brain circuits that are functionally heterogeneous at the single-cell level. While immediate early gene mapping has been widely used for decades to identify brain regions which are activated by external stimuli, recent characterization of the promoter and enhancer elements responsible for neuronal activity-dependent transcription have opened new avenues for live imaging of active neurons. Indeed, these advancements provided the basis for a growing repertoire of novel experiments to address the role of active neuronal networks in cognitive behaviors. In this review, we summarize the current literature on the usage and development of activity-dependent promoters and discuss the future directions of this expanding new field.
Collapse
Affiliation(s)
- Takashi Kawashima
- Department of Neurochemistry, Graduate School of Medicine, The University of TokyoTokyo, Japan
| | - Hiroyuki Okuno
- Department of Neurochemistry, Graduate School of Medicine, The University of TokyoTokyo, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, Graduate School of Medicine, The University of TokyoTokyo, Japan
- Core Research for Evolutionary Science and Technology, Japan Science and Technology AgencySaitama, Japan
| |
Collapse
|
46
|
In vivo imaging of immediate early gene expression reveals layer-specific memory traces in the mammalian brain. Proc Natl Acad Sci U S A 2014; 111:2788-93. [PMID: 24550309 DOI: 10.1073/pnas.1316808111] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The dynamic processes of formatting long-term memory traces in the cortex are poorly understood. The investigation of these processes requires measurements of task-evoked neuronal activities from large numbers of neurons over many days. Here, we present a two-photon imaging-based system to track event-related neuronal activity in thousands of neurons through the quantitative measurement of EGFP proteins expressed under the control of the EGR1 gene promoter. A recognition algorithm was developed to detect GFP-positive neurons in multiple cortical volumes and thereby to allow the reproducible tracking of 4,000 neurons in each volume for 2 mo. The analysis revealed a context-specific response in sparse layer II neurons. The context-evoked response gradually increased during several days of training and was maintained 1 mo later. The formed traces were specifically activated by the training context and were linearly correlated with the behavioral response. Neuronal assemblies that responded to specific contexts were largely separated, indicating the sparse coding of memory-related traces in the layer II cortical circuit.
Collapse
|
47
|
Cruz FC, Koya E, Guez-Barber DH, Bossert JM, Lupica CR, Shaham Y, Hope BT. New technologies for examining the role of neuronal ensembles in drug addiction and fear. Nat Rev Neurosci 2013; 14:743-54. [PMID: 24088811 DOI: 10.1038/nrn3597] [Citation(s) in RCA: 198] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Correlational data suggest that learned associations are encoded within neuronal ensembles. However, it has been difficult to prove that neuronal ensembles mediate learned behaviours because traditional pharmacological and lesion methods, and even newer cell type-specific methods, affect both activated and non-activated neurons. In addition, previous studies on synaptic and molecular alterations induced by learning did not distinguish between behaviourally activated and non-activated neurons. Here, we describe three new approaches--Daun02 inactivation, FACS sorting of activated neurons and Fos-GFP transgenic rats--that have been used to selectively target and study activated neuronal ensembles in models of conditioned drug effects and relapse. We also describe two new tools--Fos-tTA transgenic mice and inactivation of CREB-overexpressing neurons--that have been used to study the role of neuronal ensembles in conditioned fear.
Collapse
Affiliation(s)
- Fabio C Cruz
- Intramural Research Program, National Institute on Drug Abuse-National Institutes of Health, 251 Bayview Boulevard, Baltimore, Maryland 21224, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
On the potential role of active DNA demethylation in establishing epigenetic states associated with neural plasticity and memory. Neurobiol Learn Mem 2013; 105:125-32. [PMID: 23806749 DOI: 10.1016/j.nlm.2013.06.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 06/11/2013] [Accepted: 06/11/2013] [Indexed: 11/20/2022]
Abstract
Dynamic variations in DNA methylation regulate neuronal gene expression in an experience-dependent manner. Although DNA methylation has been implicated in synaptic plasticity, learning and memory, active DNA demethylation is also induced by learning, which suggests that an interaction between the two processes is necessary for cognitive function. Active DNA demethylation is a complex process involving a variety of proteins and epigenetic regulatory enzymes, the understanding of which with respect to its role in the adult brain is in its infancy. We here provide an overview of the current understanding of active DNA demethylation, and describe how this process may establish persistent epigenetic states that are associated with neural plasticity and memory formation.
Collapse
|
49
|
Cao VY, Ye Y, Mastwal SS, Lovinger DM, Costa RM, Wang KH. In vivo two-photon imaging of experience-dependent molecular changes in cortical neurons. J Vis Exp 2013:50148. [PMID: 23329071 DOI: 10.3791/50148] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The brain's ability to change in response to experience is essential for healthy brain function, and abnormalities in this process contribute to a variety of brain disorders. To better understand the mechanisms by which brain circuits react to an animal's experience requires the ability to monitor the experience-dependent molecular changes in a given set of neurons, over a prolonged period of time, in the live animal. While experience and associated neural activity is known to trigger gene expression changes in neurons most of the methods to detect such changes do not allow repeated observation of the same neurons over multiple days or do not have sufficient resolution to observe individual neurons. Here, we describe a method that combines in vivo two-photon microscopy with a genetically encoded fluorescent reporter to track experience-dependent gene expression changes in individual cortical neurons over the course of day-to-day experience. One of the well-established experience-dependent genes is Activity-regulated cytoskeletal associated protein (Arc). The transcription of Arc is rapidly and highly induced by intensified neuronal activity and its protein product regulates the endocytosis of glutamate receptors and long-term synaptic plasticity. The expression of Arc has been widely used as a molecular marker to map neuronal circuits involved in specific behaviors. In most of those studies, Arc expression was detected by in situ hybridization or immunohistochemistry in fixed brain sections. Although those methods revealed that the expression of Arc was localized to a subset of excitatory neurons after behavioral experience, how the cellular patterns of Arc expression might change with multiple episodes of repeated or distinctive experiences over days was not investigated. In vivo two-photon microscopy offers a powerful way to examine experience-dependent cellular changes in the living brain. To enable the examination of Arc expression in live neurons by two-photon microscopy, we previously generated a knock-in mouse line in which a GFP reporter is placed under the control of the endogenous Arc promoter. This protocol describes the surgical preparations and imaging procedures for tracking experience-dependent Arc-GFP expression patterns in neuronal ensembles in the live animal. In this method, chronic cranial windows were first implanted in Arc-GFP mice over the cortical regions of interest. Those animals were then repeatedly imaged by two-photon microscopy after desired behavioral paradigms over the course of several days. This method may be generally applicable to animals carrying other fluorescent reporters of experience-dependent molecular changes.
Collapse
Affiliation(s)
- Vania Y Cao
- Unit on Neural Circuits and Adaptive Behaviors, Genes Cognition and Psychosis Program, National Institute of Mental Health, USA
| | | | | | | | | | | |
Collapse
|
50
|
Spiacci A, Coimbra N, Zangrossi H. Differential involvement of dorsal raphe subnuclei in the regulation of anxiety- and panic-related defensive behaviors. Neuroscience 2012; 227:350-60. [DOI: 10.1016/j.neuroscience.2012.09.061] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 08/29/2012] [Accepted: 09/25/2012] [Indexed: 10/27/2022]
|