1
|
Uchigashima M, Mikuni T. Single-cell synaptome mapping: its technical basis and applications in critical period plasticity research. Front Neural Circuits 2024; 18:1523614. [PMID: 39726910 PMCID: PMC11670323 DOI: 10.3389/fncir.2024.1523614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 11/28/2024] [Indexed: 12/28/2024] Open
Abstract
Our brain adapts to the environment by optimizing its function through experience-dependent cortical plasticity. This plasticity is transiently enhanced during a developmental stage, known as the "critical period," and subsequently maintained at lower levels throughout adulthood. Thus, understanding the mechanism underlying critical period plasticity is crucial for improving brain adaptability across the lifespan. Critical period plasticity relies on activity-dependent circuit remodeling through anatomical and functional changes at individual synapses. However, it remains challenging to identify the molecular signatures of synapses responsible for critical period plasticity and to understand how these plasticity-related synapses are spatiotemporally organized within a neuron. Recent advances in genetic tools and genome editing methodologies have enabled single-cell endogenous protein labeling in the brain, allowing for comprehensive molecular profiling of individual synapses within a neuron, namely "single-cell synaptome mapping." This promising approach can facilitate insights into the spatiotemporal organization of synapses that are sparse yet functionally important within single neurons. In this review, we introduce the basics of single-cell synaptome mapping and discuss its methodologies and applications to investigate the synaptic and cellular mechanisms underlying circuit remodeling during the critical period.
Collapse
Affiliation(s)
- Motokazu Uchigashima
- Department of Cellular Neuropathology, Brain Research Institute, Niigata University, Niigata, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, Tokyo, Japan
| | - Takayasu Mikuni
- Department of Cellular Neuropathology, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
2
|
Matsui A, Spangler C, Elferich J, Shiozaki M, Jean N, Zhao X, Qin M, Zhong H, Yu Z, Gouaux E. Cryo-electron tomographic investigation of native hippocampal glutamatergic synapses. eLife 2024; 13:RP98458. [PMID: 39495821 PMCID: PMC11534335 DOI: 10.7554/elife.98458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2024] Open
Abstract
Chemical synapses are the major sites of communication between neurons in the nervous system and mediate either excitatory or inhibitory signaling. At excitatory synapses, glutamate is the primary neurotransmitter and upon release from presynaptic vesicles, is detected by postsynaptic glutamate receptors, which include ionotropic AMPA and NMDA receptors. Here, we have developed methods to identify glutamatergic synapses in brain tissue slices, label AMPA receptors with small gold nanoparticles (AuNPs), and prepare lamella for cryo-electron tomography studies. The targeted imaging of glutamatergic synapses in the lamella is facilitated by fluorescent pre- and postsynaptic signatures, and the subsequent tomograms allow for the identification of key features of chemical synapses, including synaptic vesicles, the synaptic cleft, and AuNP-labeled AMPA receptors. These methods pave the way for imaging brain regions at high resolution, using unstained, unfixed samples preserved under near-native conditions.
Collapse
Affiliation(s)
- Aya Matsui
- Howard Hughes Medical Institute, Oregon Health and Science UniversityPortlandUnited States
- Vollum Institute, Oregon Health and Science UniversityPortlandUnited States
| | - Cathy Spangler
- Vollum Institute, Oregon Health and Science UniversityPortlandUnited States
| | - Johannes Elferich
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Howard Hughes Medical InstituteWorcesterUnited States
| | - Momoko Shiozaki
- Howard Hughes Medical Institute, Janelia Research InstituteAshburnUnited States
| | - Nikki Jean
- Howard Hughes Medical Institute, Janelia Research InstituteAshburnUnited States
| | - Xiaowei Zhao
- Howard Hughes Medical Institute, Janelia Research InstituteAshburnUnited States
| | - Maozhen Qin
- Vollum Institute, Oregon Health and Science UniversityPortlandUnited States
| | - Haining Zhong
- Vollum Institute, Oregon Health and Science UniversityPortlandUnited States
| | - Zhiheng Yu
- Howard Hughes Medical Institute, Janelia Research InstituteAshburnUnited States
| | - Eric Gouaux
- Howard Hughes Medical Institute, Oregon Health and Science UniversityPortlandUnited States
- Vollum Institute, Oregon Health and Science UniversityPortlandUnited States
| |
Collapse
|
3
|
Morabito A, Zerlau Y, Dhanasobhon D, Berthaux E, Tzilivaki A, Moneron G, Cathala L, Poirazi P, Bacci A, DiGregorio D, Lourenço J, Rebola N. A dendritic substrate for temporal diversity of cortical inhibition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.09.602783. [PMID: 39026855 PMCID: PMC11257522 DOI: 10.1101/2024.07.09.602783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
In the mammalian neocortex, GABAergic interneurons (INs) inhibit cortical networks in profoundly different ways. The extent to which this depends on how different INs process excitatory signals along their dendrites is poorly understood. Here, we reveal that the functional specialization of two major populations of cortical INs is determined by the unique association of different dendritic integration modes with distinct synaptic organization motifs. We found that somatostatin (SST)-INs exhibit NMDAR-dependent dendritic integration and uniform synapse density along the dendritic tree. In contrast, dendrites of parvalbumin (PV)-INs exhibit passive synaptic integration coupled with proximally enriched synaptic distributions. Theoretical analysis shows that these two dendritic configurations result in different strategies to optimize synaptic efficacy in thin dendritic structures. Yet, the two configurations lead to distinct temporal engagement of each IN during network activity. We confirmed these predictions with in vivo recordings of IN activity in the visual cortex of awake mice, revealing a rapid and linear recruitment of PV-INs as opposed to a long-lasting integrative activation of SST-INs. Our work reveals the existence of distinct dendritic strategies that confer distinct temporal representations for the two major classes of neocortical INs and thus dynamics of inhibition.
Collapse
Affiliation(s)
- Annunziato Morabito
- ICM, Paris Brain Institute, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, Inserm, CNRS, Paris, 75013, France
| | - Yann Zerlau
- ICM, Paris Brain Institute, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, Inserm, CNRS, Paris, 75013, France
| | - Dhanasak Dhanasobhon
- ICM, Paris Brain Institute, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, Inserm, CNRS, Paris, 75013, France
| | - Emmanuelle Berthaux
- ICM, Paris Brain Institute, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, Inserm, CNRS, Paris, 75013, France
| | - Alexandra Tzilivaki
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität zu Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Neuroscience Research Center, 10117 Berlin, Germany
- Einstein Center for Neurosciences, Chariteplatz 1, 10117 Berlin, Germany
- NeuroCure Cluster of Excellence, Chariteplatz 1, 10117 Berlin, Germany
| | - Gael Moneron
- Institut Pasteur, CNRS UMR3571, Synapse and Circuit Dynamics Unit, 75015 Paris, France
| | - Laurence Cathala
- ICM, Paris Brain Institute, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, Inserm, CNRS, Paris, 75013, France
| | - Panayiota Poirazi
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology Hellas (FORTH), Heraklion, 70013, Greece
| | - Alberto Bacci
- ICM, Paris Brain Institute, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, Inserm, CNRS, Paris, 75013, France
| | - David DiGregorio
- Institut Pasteur, CNRS UMR3571, Synapse and Circuit Dynamics Unit, 75015 Paris, France
| | - Joana Lourenço
- ICM, Paris Brain Institute, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, Inserm, CNRS, Paris, 75013, France
| | - Nelson Rebola
- ICM, Paris Brain Institute, Hôpital de la Pitié-Salpêtrière, Sorbonne Université, Inserm, CNRS, Paris, 75013, France
| |
Collapse
|
4
|
Ray A, Loghinov I, Ravindranath V, Barth AL. Early hippocampal hyperexcitability and synaptic reorganization in mouse models of amyloidosis. iScience 2024; 27:110629. [PMID: 39262788 PMCID: PMC11388185 DOI: 10.1016/j.isci.2024.110629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/09/2024] [Accepted: 07/29/2024] [Indexed: 09/13/2024] Open
Abstract
The limited success of plaque-reducing therapies in Alzheimer's disease suggests that early treatment might be more effective in delaying or reversing memory impairments. Toward this end, it is important to establish the progression of synaptic and circuit changes before onset of plaques or cognitive deficits. Here, we used quantitative, fluorescence-based methods for synapse detection in CA1 pyramidal neurons to investigate the interaction between abnormal circuit activity, measured by Fos-immunoreactivity, and synapse reorganization in mouse models of amyloidosis. Using a genetically encoded, fluorescently labeled synaptic marker in juvenile mice (prior to sexual maturity), we find both synapse gain and loss depending on dendritic location. This progresses to broad synapse loss in aged mice. Elevated hippocampal activity in both CA3 and CA1 was present at weaning and preceded this reorganization. Thus, Aβ overproduction may initiate abnormal activity and subsequent input-specific synapse plasticity. These findings indicate that sustained amyloidosis drives heterogeneous and progressive circuit-wide abnormalities.
Collapse
Affiliation(s)
- Ajit Ray
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Iulia Loghinov
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Vijayalakshmi Ravindranath
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, Karnataka 560012, India
- Centre for Brain Research, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Alison L. Barth
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
5
|
McMullan HM, Gansemer BM, Thayer SA. Antiretroviral drugs from multiple classes induce loss of excitatory synapses between hippocampal neurons in culture. Front Pharmacol 2024; 15:1369757. [PMID: 38533258 PMCID: PMC10963620 DOI: 10.3389/fphar.2024.1369757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 02/27/2024] [Indexed: 03/28/2024] Open
Abstract
Introduction: Antiretroviral (ARV) drugs have improved prognoses for people living with HIV. However, HIV-associated neurocognitive disorders (HAND) persist despite undetectable viral loads. Some ARVs have been linked to neuropsychiatric effects that may contribute to HAND. Synapse loss correlates with cognitive decline in HAND and synaptic deficits may contribute to the neuropsychiatric effects of ARV drugs. Methods: Using an automated high content assay, rat hippocampal neurons in culture expressing PSD95-eGFP to label glutamatergic synapses and mCherry to fill neuronal structures were imaged before and after treatment with 25 clinically used ARVs. Results and Discussion: At a concentration of 10 μM the protease inhibitors nelfinavir and saquinavir, the non-nucleoside reverse transcriptase inhibitors etravirine and the 8-OH metabolite of efavirenz, the integrase inhibitor bictegravir, and the capsid inhibitor lenacapavir produced synaptic toxicity. Only lenacapavir produced synapse loss at the nanomolar concentrations estimated free in the plasma, although all 4 ARV drugs induced synapse loss at Cmax. Evaluation of combination therapies did not reveal synergistic synaptic toxicity. Synapse loss developed fully by 24 h and persisted for at least 3 days. Bictegravir-induced synapse loss required activation of voltage-gated Ca2+ channels and bictegravir, etravirine, and lenacapavir produced synapse loss by an excitotoxic mechanism. These results indicate that select ARV drugs might contribute to neuropsychiatric effects in combination with drugs that bind serum proteins or in disease states in which synaptic function is altered. The high content imaging assay used here provides an efficient means to evaluate new drugs and drug combinations for potential CNS toxicity.
Collapse
Affiliation(s)
| | | | - Stanley A. Thayer
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
6
|
Li J, Miramontes TG, Czopka T, Monk KR. Synaptic input and Ca 2+ activity in zebrafish oligodendrocyte precursor cells contribute to myelin sheath formation. Nat Neurosci 2024; 27:219-231. [PMID: 38216650 DOI: 10.1038/s41593-023-01553-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 12/13/2023] [Indexed: 01/14/2024]
Abstract
In the nervous system, only one type of neuron-glial synapse is known to exist: that between neurons and oligodendrocyte precursor cells (OPCs), yet their composition, assembly, downstream signaling and in vivo functions remain largely unclear. Here, we address these questions using in vivo microscopy in zebrafish spinal cord and identify postsynaptic molecules PSD-95 and gephyrin in OPCs. The puncta containing these molecules in OPCs increase during early development and decrease upon OPC differentiation. These puncta are highly dynamic and frequently assemble at 'hotspots'. Gephyrin hotspots and synapse-associated Ca2+ activity in OPCs predict where a subset of myelin sheaths forms in differentiated oligodendrocytes. Further analyses reveal that spontaneous synaptic release is integral to OPC Ca2+ activity, while evoked synaptic release contributes only in early development. Finally, disruption of the synaptic genes dlg4a/dlg4b, gphnb and nlgn3b impairs OPC differentiation and myelination. Together, we propose that neuron-OPC synapses are dynamically assembled and can predetermine myelination patterns through Ca2+ signaling.
Collapse
Affiliation(s)
- Jiaxing Li
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA.
| | | | - Tim Czopka
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Kelly R Monk
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
7
|
Niraula S, Yan SS, Subramanian J. Amyloid Pathology Impairs Experience-Dependent Inhibitory Synaptic Plasticity. J Neurosci 2024; 44:e0702232023. [PMID: 38050105 PMCID: PMC10860629 DOI: 10.1523/jneurosci.0702-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 11/09/2023] [Accepted: 11/14/2023] [Indexed: 12/06/2023] Open
Abstract
Alzheimer's disease patients and mouse models exhibit aberrant neuronal activity and altered excitatory-to-inhibitory synaptic ratio. Using multicolor two-photon microscopy, we test how amyloid pathology alters the structural dynamics of excitatory and inhibitory synapses and their adaptation to altered visual experience in vivo in the visual cortex. We show that the baseline dynamics of mature excitatory synapses and their adaptation to visual deprivation are not altered in amyloidosis. Likewise, the baseline dynamics of inhibitory synapses are not affected. In contrast, visual deprivation fails to induce inhibitory synapse loss in amyloidosis, a phenomenon observed in nonpathological conditions. Intriguingly, inhibitory synapse loss associated with visual deprivation in nonpathological mice is accompanied by subtle broadening of spontaneous but not visually evoked calcium transients. However, such broadening does not manifest in the context of amyloidosis. We also show that excitatory and inhibitory synapse loss is locally clustered under the nonpathological state. In contrast, a fraction of synapse loss is not locally clustered in amyloidosis, indicating an impairment in inhibitory synapse adaptation to changes in excitatory synaptic activity.
Collapse
Affiliation(s)
- Suraj Niraula
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, Kansas 66045
| | - Shirley ShiDu Yan
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, Kansas 66045
- Department of Surgery, Vagelos College of Physicians and Surgeons of Columbia University, New York, New York 10032
| | - Jaichandar Subramanian
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, Kansas 66045
| |
Collapse
|
8
|
Rimbault C, Breillat C, Compans B, Toulmé E, Vicente FN, Fernandez-Monreal M, Mascalchi P, Genuer C, Puente-Muñoz V, Gauthereau I, Hosy E, Claverol S, Giannone G, Chamma I, Mackereth CD, Poujol C, Choquet D, Sainlos M. Engineering paralog-specific PSD-95 recombinant binders as minimally interfering multimodal probes for advanced imaging techniques. eLife 2024; 13:e69620. [PMID: 38167295 PMCID: PMC10803022 DOI: 10.7554/elife.69620] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
Despite the constant advances in fluorescence imaging techniques, monitoring endogenous proteins still constitutes a major challenge in particular when considering dynamics studies or super-resolution imaging. We have recently evolved specific protein-based binders for PSD-95, the main postsynaptic scaffold proteins at excitatory synapses. Since the synthetic recombinant binders recognize epitopes not directly involved in the target protein activity, we consider them here as tools to develop endogenous PSD-95 imaging probes. After confirming their lack of impact on PSD-95 function, we validated their use as intrabody fluorescent probes. We further engineered the probes and demonstrated their usefulness in different super-resolution imaging modalities (STED, PALM, and DNA-PAINT) in both live and fixed neurons. Finally, we exploited the binders to enrich at the synapse genetically encoded calcium reporters. Overall, we demonstrate that these evolved binders constitute a robust and efficient platform to selectively target and monitor endogenous PSD-95 using various fluorescence imaging techniques.
Collapse
Affiliation(s)
- Charlotte Rimbault
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297BordeauxFrance
| | - Christelle Breillat
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297BordeauxFrance
| | - Benjamin Compans
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297BordeauxFrance
| | - Estelle Toulmé
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297BordeauxFrance
| | - Filipe Nunes Vicente
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297BordeauxFrance
| | - Monica Fernandez-Monreal
- University of Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4BordeauxFrance
| | - Patrice Mascalchi
- University of Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4BordeauxFrance
| | - Camille Genuer
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297BordeauxFrance
| | - Virginia Puente-Muñoz
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297BordeauxFrance
| | - Isabel Gauthereau
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297BordeauxFrance
| | - Eric Hosy
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297BordeauxFrance
| | | | - Gregory Giannone
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297BordeauxFrance
| | - Ingrid Chamma
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297BordeauxFrance
| | | | - Christel Poujol
- University of Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, US 4BordeauxFrance
| | - Daniel Choquet
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297BordeauxFrance
| | - Matthieu Sainlos
- University of Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297BordeauxFrance
| |
Collapse
|
9
|
Bygrave AM, Sengupta A, Jackert EP, Ahmed M, Adenuga B, Nelson E, Goldschmidt HL, Johnson RC, Zhong H, Yeh FL, Sheng M, Huganir RL. Btbd11 supports cell-type-specific synaptic function. Cell Rep 2023; 42:112591. [PMID: 37261953 PMCID: PMC10592477 DOI: 10.1016/j.celrep.2023.112591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 04/21/2023] [Accepted: 05/16/2023] [Indexed: 06/03/2023] Open
Abstract
Synapses in the brain exhibit cell-type-specific differences in basal synaptic transmission and plasticity. Here, we evaluated cell-type-specific specializations in the composition of glutamatergic synapses, identifying Btbd11 as an inhibitory interneuron-specific, synapse-enriched protein. Btbd11 is highly conserved across species and binds to core postsynaptic proteins, including Psd-95. Intriguingly, we show that Btbd11 can undergo liquid-liquid phase separation when expressed with Psd-95, supporting the idea that the glutamatergic postsynaptic density in synapses in inhibitory interneurons exists in a phase-separated state. Knockout of Btbd11 decreased glutamatergic signaling onto parvalbumin-positive interneurons. Further, both in vitro and in vivo, Btbd11 knockout disrupts network activity. At the behavioral level, Btbd11 knockout from interneurons alters exploratory behavior, measures of anxiety, and sensitizes mice to pharmacologically induced hyperactivity following NMDA receptor antagonist challenge. Our findings identify a cell-type-specific mechanism that supports glutamatergic synapse function in inhibitory interneurons-with implications for circuit function and animal behavior.
Collapse
Affiliation(s)
- Alexei M Bygrave
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Ayesha Sengupta
- National Institute on Drug Abuse, Bayview Boulevard, Baltimore, MD 21224, USA
| | - Ella P Jackert
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Mehroz Ahmed
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Beloved Adenuga
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Erik Nelson
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hana L Goldschmidt
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Richard C Johnson
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Haining Zhong
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Felix L Yeh
- Department of Neuroscience, Genentech, Inc, South San Francisco, CA 94080, USA
| | - Morgan Sheng
- Department of Neuroscience, Genentech, Inc, South San Francisco, CA 94080, USA
| | - Richard L Huganir
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
10
|
Kilisch M, Gere-Becker M, Wüstefeld L, Bonnas C, Crauel A, Mechmershausen M, Martens H, Götzke H, Opazo F, Frey S. Simple and Highly Efficient Detection of PSD95 Using a Nanobody and Its Recombinant Heavy-Chain Antibody Derivatives. Int J Mol Sci 2023; 24:ijms24087294. [PMID: 37108454 PMCID: PMC10138605 DOI: 10.3390/ijms24087294] [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: 03/16/2023] [Revised: 04/03/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
The post-synaptic density protein 95 (PSD95) is a crucial scaffolding protein participating in the organization and regulation of synapses. PSD95 interacts with numerous molecules, including neurotransmitter receptors and ion channels. The functional dysregulation of PSD95 as well as its abundance and localization has been implicated with several neurological disorders, making it an attractive target for developing strategies able to monitor PSD95 accurately for diagnostics and therapeutics. This study characterizes a novel camelid single-domain antibody (nanobody) that binds strongly and with high specificity to rat, mouse, and human PSD95. This nanobody allows for more precise detection and quantification of PSD95 in various biological samples. We expect that the flexibility and unique performance of this thoroughly characterized affinity tool will help to further understand the role of PSD95 in normal and diseased neuronal synapses.
Collapse
Affiliation(s)
- Markus Kilisch
- NanoTag Biotechnologies GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| | - Maja Gere-Becker
- NanoTag Biotechnologies GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| | - Liane Wüstefeld
- Synaptic Systems GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| | - Christel Bonnas
- Synaptic Systems GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| | - Alexander Crauel
- NanoTag Biotechnologies GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| | - Maja Mechmershausen
- NanoTag Biotechnologies GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| | - Henrik Martens
- Synaptic Systems GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| | - Hansjörg Götzke
- NanoTag Biotechnologies GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| | - Felipe Opazo
- NanoTag Biotechnologies GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany
- Center for Biostructural Imaging of Neurodegeneration (BIN), University of Göttingen Medical Center, 37075 Göttingen, Germany
| | - Steffen Frey
- NanoTag Biotechnologies GmbH, Rudolf-Wissell-Straβe 28a, 37079 Göttingen, Germany
| |
Collapse
|
11
|
Droogers WJ, MacGillavry HD. Plasticity of postsynaptic nanostructure. Mol Cell Neurosci 2023; 124:103819. [PMID: 36720293 DOI: 10.1016/j.mcn.2023.103819] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 01/31/2023] Open
Abstract
The postsynaptic density (PSD) of excitatory synapses is built from a wide variety of scaffolding proteins, receptors, and signaling molecules that collectively orchestrate synaptic transmission. Seminal work over the past decades has led to the identification and functional characterization of many PSD components. In contrast, we know far less about how these constituents are assembled within synapses, and how this organization contributes to synapse function. Notably, recent evidence from high-resolution microscopy studies and in silico models, highlights the importance of the precise subsynaptic structure of the PSD for controlling the strength of synaptic transmission. Even further, activity-driven changes in the distribution of glutamate receptors are acknowledged to contribute to long-term changes in synaptic efficacy. Thus, defining the mechanisms that drive structural changes within the PSD are important for a molecular understanding of synaptic transmission and plasticity. Here, we review the current literature on how the PSD is organized to mediate basal synaptic transmission and how synaptic activity alters the nanoscale organization of synapses to sustain changes in synaptic strength.
Collapse
Affiliation(s)
- W J Droogers
- Division of Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 CH, The Netherlands
| | - H D MacGillavry
- Division of Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, 3584 CH, The Netherlands.
| |
Collapse
|
12
|
Ray A, Christian JA, Mosso MB, Park E, Wegner W, Willig KI, Barth AL. Quantitative Fluorescence Analysis Reveals Dendrite-Specific Thalamocortical Plasticity in L5 Pyramidal Neurons during Learning. J Neurosci 2023; 43:584-600. [PMID: 36639912 PMCID: PMC9888508 DOI: 10.1523/jneurosci.1372-22.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/28/2022] [Accepted: 11/23/2022] [Indexed: 12/14/2022] Open
Abstract
High-throughput anatomic data can stimulate and constrain new hypotheses about how neural circuits change in response to experience. Here, we use fluorescence-based reagents for presynaptic and postsynaptic labeling to monitor changes in thalamocortical synapses onto different compartments of layer 5 (L5) pyramidal (Pyr) neurons in somatosensory (barrel) cortex from mixed-sex mice during whisker-dependent learning (Audette et al., 2019). Using axonal fills and molecular-genetic tags for synapse identification in fixed tissue from Rbp4-Cre transgenic mice, we found that thalamocortical synapses from the higher-order posterior medial thalamic nucleus showed rapid morphologic changes in both presynaptic and postsynaptic structures at the earliest stages of sensory association training. Detected increases in thalamocortical synaptic size were compartment specific, occurring selectively in the proximal dendrites onto L5 Pyr and not at inputs onto their apical tufts in L1. Both axonal and dendritic changes were transient, normalizing back to baseline as animals became expert in the task. Anatomical measurements were corroborated by electrophysiological recordings at different stages of training. Thus, fluorescence-based analysis of input- and target-specific synapses can reveal compartment-specific changes in synapse properties during learning.SIGNIFICANCE STATEMENT Synaptic changes underlie the cellular basis of learning, experience, and neurologic diseases. Neuroanatomical methods to assess synaptic plasticity can provide critical spatial information necessary for building models of neuronal computations during learning and experience but are technically and fiscally intensive. Here, we describe a confocal fluorescence microscopy-based analytical method to assess input, cell type, and dendritic location-specific synaptic plasticity in a sensory learning assay. Our method not only confirms prior electrophysiological measurements but allows us to predict functional strength of synapses in a pathway-specific manner. Our findings also indicate that changes in primary sensory cortices are transient, occurring during early learning. Fluorescence-based synapse identification can be an efficient and easily adopted approach to study synaptic changes in a variety of experimental paradigms.
Collapse
Affiliation(s)
- Ajit Ray
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh Pennsylvania 15213
| | - Joseph A Christian
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh Pennsylvania 15213
| | - Matthew B Mosso
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh Pennsylvania 15213
| | - Eunsol Park
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh Pennsylvania 15213
| | - Waja Wegner
- Optical Nanoscopy in Neuroscience, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, 37099 Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany
| | - Katrin I Willig
- Optical Nanoscopy in Neuroscience, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, 37099 Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany
| | - Alison L Barth
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh Pennsylvania 15213
| |
Collapse
|
13
|
Bulovaite E, Qiu Z, Kratschke M, Zgraj A, Fricker DG, Tuck EJ, Gokhale R, Koniaris B, Jami SA, Merino-Serrais P, Husi E, Mendive-Tapia L, Vendrell M, O'Dell TJ, DeFelipe J, Komiyama NH, Holtmaat A, Fransén E, Grant SGN. A brain atlas of synapse protein lifetime across the mouse lifespan. Neuron 2022; 110:4057-4073.e8. [PMID: 36202095 PMCID: PMC9789179 DOI: 10.1016/j.neuron.2022.09.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 07/01/2022] [Accepted: 09/07/2022] [Indexed: 11/12/2022]
Abstract
The lifetime of proteins in synapses is important for their signaling, maintenance, and remodeling, and for memory duration. We quantified the lifetime of endogenous PSD95, an abundant postsynaptic protein in excitatory synapses, at single-synapse resolution across the mouse brain and lifespan, generating the Protein Lifetime Synaptome Atlas. Excitatory synapses have a wide range of PSD95 lifetimes extending from hours to several months, with distinct spatial distributions in dendrites, neurons, and brain regions. Synapses with short protein lifetimes are enriched in young animals and in brain regions controlling innate behaviors, whereas synapses with long protein lifetimes accumulate during development, are enriched in the cortex and CA1 where memories are stored, and are preferentially preserved in old age. Synapse protein lifetime increases throughout the brain in a mouse model of autism and schizophrenia. Protein lifetime adds a further layer to synapse diversity and enriches prevailing concepts in brain development, aging, and disease.
Collapse
Affiliation(s)
- Edita Bulovaite
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Zhen Qiu
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Maximilian Kratschke
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Adrianna Zgraj
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - David G Fricker
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Eleanor J Tuck
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Ragini Gokhale
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Babis Koniaris
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; School of Computing, Edinburgh Napier University, Edinburgh EH10 5DT, UK
| | - Shekib A Jami
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Integrative Center for Learning and Memory, Brain Research Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Paula Merino-Serrais
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, UPM, 28223 Madrid, Spain; Instituto Cajal, CSIC, 28002 Madrid, Spain
| | - Elodie Husi
- Department of Basic Neurosciences, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Lorena Mendive-Tapia
- Centre for Inflammation Research, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Marc Vendrell
- Centre for Inflammation Research, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Thomas J O'Dell
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Integrative Center for Learning and Memory, Brain Research Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Javier DeFelipe
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, UPM, 28223 Madrid, Spain; Instituto Cajal, CSIC, 28002 Madrid, Spain
| | - Noboru H Komiyama
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; Simons Initiative for the Developing Brain (SIDB), Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK; The Patrick Wild Centre for Research into Autism, Fragile X Syndrome & Intellectual Disabilities, Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK; Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Anthony Holtmaat
- Department of Basic Neurosciences, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland
| | - Erik Fransén
- Department of Computational Science and Technology, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, 10044 Stockholm, Sweden; Science for Life Laboratory, KTH Royal Institute of Technology, 171 65 Solna, Sweden
| | - Seth G N Grant
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; Simons Initiative for the Developing Brain (SIDB), Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK.
| |
Collapse
|
14
|
Qi C, Luo LD, Feng I, Ma S. Molecular mechanisms of synaptogenesis. Front Synaptic Neurosci 2022; 14:939793. [PMID: 36176941 PMCID: PMC9513053 DOI: 10.3389/fnsyn.2022.939793] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/27/2022] [Indexed: 11/29/2022] Open
Abstract
Synapses are the basic units for information processing and storage in the nervous system. It is only when the synaptic connection is established, that it becomes meaningful to discuss the structure and function of a circuit. In humans, our unparalleled cognitive abilities are correlated with an increase in the number of synapses. Additionally, genes involved in synaptogenesis are also frequently associated with neurological or psychiatric disorders, suggesting a relationship between synaptogenesis and brain physiology and pathology. Thus, understanding the molecular mechanisms of synaptogenesis is the key to the mystery of circuit assembly and neural computation. Furthermore, it would provide therapeutic insights for the treatment of neurological and psychiatric disorders. Multiple molecular events must be precisely coordinated to generate a synapse. To understand the molecular mechanisms underlying synaptogenesis, we need to know the molecular components of synapses, how these molecular components are held together, and how the molecular networks are refined in response to neural activity to generate new synapses. Thanks to the intensive investigations in this field, our understanding of the process of synaptogenesis has progressed significantly. Here, we will review the molecular mechanisms of synaptogenesis by going over the studies on the identification of molecular components in synapses and their functions in synaptogenesis, how cell adhesion molecules connect these synaptic molecules together, and how neural activity mobilizes these molecules to generate new synapses. Finally, we will summarize the human-specific regulatory mechanisms in synaptogenesis and results from human genetics studies on synaptogenesis and brain disorders.
Collapse
Affiliation(s)
- Cai Qi
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, United States
- *Correspondence: Cai Qi,
| | - Li-Da Luo
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, United States
- Department of Cellular and Molecular Physiology, Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, United States
| | - Irena Feng
- Boston University School of Medicine, Boston, MA, United States
| | - Shaojie Ma
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
15
|
Wilson EA, Mao T, Zhong H. Labeling Endogenous Proteins Using CRISPR-mediated Insertion of Exon (CRISPIE). Bio Protoc 2022; 12:e4343. [PMID: 35592602 PMCID: PMC8918219 DOI: 10.21769/bioprotoc.4343] [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: 01/06/2022] [Revised: 10/11/2021] [Accepted: 01/13/2022] [Indexed: 12/29/2022] Open
Abstract
The CRISPR/Cas9 technology has transformed our ability to edit eukaryotic genomes. Despite this breakthrough, it remains challenging to precisely knock-in large DNA sequences, such as those encoding a fluorescent protein, for labeling or modifying a target protein in post-mitotic cells. Previous efforts focusing on sequence insertion to the protein coding sequence often suffer from insertions/deletions (INDELs) resulting from the efficient non-homologous end joining pathway (NHEJ). To overcome this limitation, we have developed CRISPR-mediated insertion of exon (CRISPIE). CRISPIE circumvents INDELs and other editing errors by inserting a designer exon flanked by adjacent intron sequences into an appropriate intronic location of the targeted gene. Because INDELs at the insertion junction can be spliced out, "CRISPIEd" genes produce precisely edited mRNA transcripts that are virtually error-free. In part due to the elimination of INDELs, high-efficiency labeling can be achieved in vivo. CRISPIE is compatible with both N- and C-terminal labels, and with all common transfection methods. Importantly, CRISPIE allows for later removal of the protein modification by including exogenous single-guide RNA (sgRNA) sites in the intronic region of the donor module. This protocol provides the detailed CRISPIE methodology, using endogenous labeling of β-actin in human U-2 OS cells with enhanced green fluorescent protein (EGFP) as an example. When combined with the appropriate gene delivery methods, the same methodology can be applied to label post-mitotic neurons in culture and in vivo. This methodology can also be readily adapted for use in other gene editing contexts.
Collapse
Affiliation(s)
- Evan A. Wilson
- Vollum Institute, Oregon Health & Sciences University, Portland, OR, USA
| | - Tianyi Mao
- Vollum Institute, Oregon Health & Sciences University, Portland, OR, USA
| | - Haining Zhong
- Vollum Institute, Oregon Health & Sciences University, Portland, OR, USA,
*For correspondence:
| |
Collapse
|
16
|
Dempsey WP, Du Z, Nadtochiy A, Smith CD, Czajkowski K, Andreev A, Robson DN, Li JM, Applebaum S, Truong TV, Kesselman C, Fraser SE, Arnold DB. Regional synapse gain and loss accompany memory formation in larval zebrafish. Proc Natl Acad Sci U S A 2022; 119:e2107661119. [PMID: 35031564 PMCID: PMC8784156 DOI: 10.1073/pnas.2107661119] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 12/03/2021] [Indexed: 11/18/2022] Open
Abstract
Defining the structural and functional changes in the nervous system underlying learning and memory represents a major challenge for modern neuroscience. Although changes in neuronal activity following memory formation have been studied [B. F. Grewe et al., Nature 543, 670-675 (2017); M. T. Rogan, U. V. Stäubli, J. E. LeDoux, Nature 390, 604-607 (1997)], the underlying structural changes at the synapse level remain poorly understood. Here, we capture synaptic changes in the midlarval zebrafish brain that occur during associative memory formation by imaging excitatory synapses labeled with recombinant probes using selective plane illumination microscopy. Imaging the same subjects before and after classical conditioning at single-synapse resolution provides an unbiased mapping of synaptic changes accompanying memory formation. In control animals and animals that failed to learn the task, there were no significant changes in the spatial patterns of synapses in the pallium, which contains the equivalent of the mammalian amygdala and is essential for associative learning in teleost fish [M. Portavella, J. P. Vargas, B. Torres, C. Salas, Brain Res. Bull 57, 397-399 (2002)]. In zebrafish that formed memories, we saw a dramatic increase in the number of synapses in the ventrolateral pallium, which contains neurons active during memory formation and retrieval. Concurrently, synapse loss predominated in the dorsomedial pallium. Surprisingly, we did not observe significant changes in the intensity of synaptic labeling, a proxy for synaptic strength, with memory formation in any region of the pallium. Our results suggest that memory formation due to classical conditioning is associated with reciprocal changes in synapse numbers in the pallium.
Collapse
Affiliation(s)
- William P Dempsey
- Department of Biological Sciences, Division of Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089
| | - Zhuowei Du
- Department of Biological Sciences, Division of Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089
| | - Anna Nadtochiy
- Translational Imaging Center, University of Southern California, Los Angeles, CA 90089
| | - Colton D Smith
- Department of Biological Sciences, Division of Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089
| | - Karl Czajkowski
- Information Sciences Institute, University of Southern California, Los Angeles, CA 90292
| | - Andrey Andreev
- Translational Imaging Center, University of Southern California, Los Angeles, CA 90089
| | - Drew N Robson
- Systems Neuroscience & Neuroengineering, Max Planck Institute for Biological Cybernetics, 72076 Tübingen, Germany
| | - Jennifer M Li
- Systems Neuroscience & Neuroengineering, Max Planck Institute for Biological Cybernetics, 72076 Tübingen, Germany
| | - Serina Applebaum
- Department of Biological Sciences, Division of Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089
| | - Thai V Truong
- Translational Imaging Center, University of Southern California, Los Angeles, CA 90089
| | - Carl Kesselman
- Information Sciences Institute, University of Southern California, Los Angeles, CA 90292
- Daniel J. Epstein Department of Industrial and Systems Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089
| | - Scott E Fraser
- Department of Biological Sciences, Division of Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089
- Translational Imaging Center, University of Southern California, Los Angeles, CA 90089
| | - Don B Arnold
- Department of Biological Sciences, Division of Molecular and Computational Biology, University of Southern California, Los Angeles, CA 90089;
| |
Collapse
|
17
|
Sneve MA, Piatkevich KD. Towards a Comprehensive Optical Connectome at Single Synapse Resolution via Expansion Microscopy. Front Synaptic Neurosci 2022; 13:754814. [PMID: 35115916 PMCID: PMC8803729 DOI: 10.3389/fnsyn.2021.754814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 12/17/2021] [Indexed: 12/04/2022] Open
Abstract
Mapping and determining the molecular identity of individual synapses is a crucial step towards the comprehensive reconstruction of neuronal circuits. Throughout the history of neuroscience, microscopy has been a key technology for mapping brain circuits. However, subdiffraction size and high density of synapses in brain tissue make this process extremely challenging. Electron microscopy (EM), with its nanoscale resolution, offers one approach to this challenge yet comes with many practical limitations, and to date has only been used in very small samples such as C. elegans, tadpole larvae, fruit fly brain, or very small pieces of mammalian brain tissue. Moreover, EM datasets require tedious data tracing. Light microscopy in combination with tissue expansion via physical magnification-known as expansion microscopy (ExM)-offers an alternative approach to this problem. ExM enables nanoscale imaging of large biological samples, which in combination with multicolor neuronal and synaptic labeling offers the unprecedented capability to trace and map entire neuronal circuits in fully automated mode. Recent advances in new methods for synaptic staining as well as new types of optical molecular probes with superior stability, specificity, and brightness provide new modalities for studying brain circuits. Here we review advanced methods and molecular probes for fluorescence staining of the synapses in the brain that are compatible with currently available expansion microscopy techniques. In particular, we will describe genetically encoded probes for synaptic labeling in mice, zebrafish, Drosophila fruit flies, and C. elegans, which enable the visualization of post-synaptic scaffolds and receptors, presynaptic terminals and vesicles, and even a snapshot of the synaptic activity itself. We will address current methods for applying these probes in ExM experiments, as well as appropriate vectors for the delivery of these molecular constructs. In addition, we offer experimental considerations and limitations for using each of these tools as well as our perspective on emerging tools.
Collapse
Affiliation(s)
- Madison A. Sneve
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, United States
| | - Kiryl D. Piatkevich
- School of Life Sciences, Westlake University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Westlake University, Hangzhou, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| |
Collapse
|
18
|
Jiang H, Esparza TJ, Kummer TT, Brody DL. Unbiased high-content screening reveals Aβ- and tau-independent synaptotoxic activities in human brain homogenates from Alzheimer's patients and high-pathology controls. PLoS One 2021; 16:e0259335. [PMID: 34748596 PMCID: PMC8575250 DOI: 10.1371/journal.pone.0259335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/19/2021] [Indexed: 11/22/2022] Open
Abstract
Alzheimer’s disease (AD) is tightly correlated with synapse loss in vulnerable brain regions. It is assumed that specific molecular entities such as Aβ and tau cause synapse loss in AD, yet unbiased screens for synaptotoxic activities have not been performed. Here, we performed size exclusion chromatography on soluble human brain homogenates from AD cases, high pathology non-demented controls, and low pathology age-matched controls using our novel high content primary cultured neuron-based screening assay. Both presynaptic and postsynaptic toxicities were elevated in homogenates from AD cases and high pathology non-demented controls to a similar extent, with more modest synaptotoxic activities in homogenates from low pathology normal controls. Surprisingly, synaptotoxic activities were found in size fractions peaking between the 17–44 kDa size standards that did not match well with Aβ and tau immunoreactive species in these homogenates. The fractions containing previously identified high molecular weight soluble amyloid beta aggregates/”oligomers” were non-toxic in this assay. Furthermore, immunodepletion of Aβ and tau did not reduce synaptotoxic activity. This result contrasts with previous findings involving the same methods applied to 3xTg-AD mouse brain extracts. The nature of the synaptotoxic species has not been identified. Overall, our data indicates one or more potential Aβ and tau independent synaptotoxic activities in human AD brain homogenates. This result aligns well with the key role of synaptic loss in the early cognitive decline and may provide new insight into AD pathophysiology.
Collapse
Affiliation(s)
- Hao Jiang
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Thomas J. Esparza
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, United States of America
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States of America
| | - Terrance T. Kummer
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - David L. Brody
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri, United States of America
- National Institute of Neurological Disorders and Stroke, Bethesda, Maryland, United States of America
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
19
|
Melander JB, Nayebi A, Jongbloets BC, Fortin DA, Qin M, Ganguli S, Mao T, Zhong H. Distinct in vivo dynamics of excitatory synapses onto cortical pyramidal neurons and parvalbumin-positive interneurons. Cell Rep 2021; 37:109972. [PMID: 34758304 PMCID: PMC8631347 DOI: 10.1016/j.celrep.2021.109972] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 09/03/2021] [Accepted: 10/19/2021] [Indexed: 11/18/2022] Open
Abstract
Cortical function relies on the balanced activation of excitatory and inhibitory neurons. However, little is known about the organization and dynamics of shaft excitatory synapses onto cortical inhibitory interneurons. Here, we use the excitatory postsynaptic marker PSD-95, fluorescently labeled at endogenous levels, as a proxy for excitatory synapses onto layer 2/3 pyramidal neurons and parvalbumin-positive (PV+) interneurons in the barrel cortex of adult mice. Longitudinal in vivo imaging under baseline conditions reveals that, although synaptic weights in both neuronal types are log-normally distributed, synapses onto PV+ neurons are less heterogeneous and more stable. Markov model analyses suggest that the synaptic weight distribution is set intrinsically by ongoing cell-type-specific dynamics, and substantial changes are due to accumulated gradual changes. Synaptic weight dynamics are multiplicative, i.e., changes scale with weights, although PV+ synapses also exhibit an additive component. These results reveal that cell-type-specific processes govern cortical synaptic strengths and dynamics. Melander et al. use a genetic strategy to visualize excitatory neuronal connections that cannot be inferred from morphology, and they monitor how the connections change over weeks in mice. They find distinct characteristics between synapses onto cells that “suppress” brain activity and those onto cells that “excite” brain activity.
Collapse
Affiliation(s)
- Joshua B Melander
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA; Neurosciences PhD Program, Stanford University, Stanford, CA 94305, USA
| | - Aran Nayebi
- Neurosciences PhD Program, Stanford University, Stanford, CA 94305, USA
| | - Bart C Jongbloets
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Dale A Fortin
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Maozhen Qin
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Surya Ganguli
- Department of Applied Physics, Stanford University, Stanford, CA 94305, USA.
| | - Tianyi Mao
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA.
| | - Haining Zhong
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
20
|
Biane C, Rückerl F, Abrahamsson T, Saint-Cloment C, Mariani J, Shigemoto R, DiGregorio DA, Sherrard RM, Cathala L. Developmental emergence of two-stage nonlinear synaptic integration in cerebellar interneurons. eLife 2021; 10:65954. [PMID: 34730085 PMCID: PMC8565927 DOI: 10.7554/elife.65954] [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: 12/20/2020] [Accepted: 09/28/2021] [Indexed: 11/13/2022] Open
Abstract
Synaptic transmission, connectivity, and dendritic morphology mature in parallel during brain development and are often disrupted in neurodevelopmental disorders. Yet how these changes influence the neuronal computations necessary for normal brain function are not well understood. To identify cellular mechanisms underlying the maturation of synaptic integration in interneurons, we combined patch-clamp recordings of excitatory inputs in mouse cerebellar stellate cells (SCs), three-dimensional reconstruction of SC morphology with excitatory synapse location, and biophysical modeling. We found that postnatal maturation of postsynaptic strength was homogeneously reduced along the somatodendritic axis, but dendritic integration was always sublinear. However, dendritic branching increased without changes in synapse density, leading to a substantial gain in distal inputs. Thus, changes in synapse distribution, rather than dendrite cable properties, are the dominant mechanism underlying the maturation of neuronal computation. These mechanisms favor the emergence of a spatially compartmentalized two-stage integration model promoting location-dependent integration within dendritic subunits.
Collapse
Affiliation(s)
- Celia Biane
- Sorbonne Université et CNRS UMR 8256, Adaptation Biologique et Vieillissement, Paris, France
| | - Florian Rückerl
- Institut Pasteur, Université de Paris, CNRS UMR 3571, Unit of Synapse and Circuit Dynamics, Paris, France
| | - Therese Abrahamsson
- Institut Pasteur, Université de Paris, CNRS UMR 3571, Unit of Synapse and Circuit Dynamics, Paris, France
| | - Cécile Saint-Cloment
- Institut Pasteur, Université de Paris, CNRS UMR 3571, Unit of Synapse and Circuit Dynamics, Paris, France
| | - Jean Mariani
- Sorbonne Université et CNRS UMR 8256, Adaptation Biologique et Vieillissement, Paris, France
| | - Ryuichi Shigemoto
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - David A DiGregorio
- Institut Pasteur, Université de Paris, CNRS UMR 3571, Unit of Synapse and Circuit Dynamics, Paris, France
| | - Rachel M Sherrard
- Sorbonne Université et CNRS UMR 8256, Adaptation Biologique et Vieillissement, Paris, France
| | - Laurence Cathala
- Sorbonne Université et CNRS UMR 8256, Adaptation Biologique et Vieillissement, Paris, France.,Paris Brain Institute, CNRS UMR 7225 - Inserm U1127 - Sorbonne Université Groupe Hospitalier Pitié Salpêtrière, Paris, France
| |
Collapse
|
21
|
Kuljis DA, Micheva KD, Ray A, Wegner W, Bowman R, Madison DV, Willig KI, Barth AL. Gephyrin-Lacking PV Synapses on Neocortical Pyramidal Neurons. Int J Mol Sci 2021; 22:ijms221810032. [PMID: 34576197 PMCID: PMC8467468 DOI: 10.3390/ijms221810032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/01/2021] [Accepted: 09/07/2021] [Indexed: 11/26/2022] Open
Abstract
Gephyrin has long been thought of as a master regulator for inhibitory synapses, acting as a scaffold to organize γ-aminobutyric acid type A receptors (GABAARs) at the post-synaptic density. Accordingly, gephyrin immunostaining has been used as an indicator of inhibitory synapses; despite this, the pan-synaptic localization of gephyrin to specific classes of inhibitory synapses has not been demonstrated. Genetically encoded fibronectin intrabodies generated with mRNA display (FingRs) against gephyrin (Gephyrin.FingR) reliably label endogenous gephyrin, and can be tagged with fluorophores for comprehensive synaptic quantitation and monitoring. Here we investigated input- and target-specific localization of gephyrin at a defined class of inhibitory synapse, using Gephyrin.FingR proteins tagged with EGFP in brain tissue from transgenic mice. Parvalbumin-expressing (PV) neuron presynaptic boutons labeled using Cre- dependent synaptophysin-tdTomato were aligned with postsynaptic Gephyrin.FingR puncta. We discovered that more than one-third of PV boutons adjacent to neocortical pyramidal (Pyr) cell somas lack postsynaptic gephyrin labeling. This finding was confirmed using correlative fluorescence and electron microscopy. Our findings suggest some inhibitory synapses may lack gephyrin. Gephyrin-lacking synapses may play an important role in dynamically regulating cell activity under different physiological conditions.
Collapse
Affiliation(s)
- Dika A. Kuljis
- Center for the Neural Basis of Cognition, Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (D.A.K.); (A.R.); (R.B.)
| | - Kristina D. Micheva
- Department of Molecular and Cellular Physiology, Stanford University, Palo Alto, CA 94304, USA; (K.D.M.); (D.V.M.)
| | - Ajit Ray
- Center for the Neural Basis of Cognition, Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (D.A.K.); (A.R.); (R.B.)
| | - Waja Wegner
- Optical Nanoscopy in Neuroscience, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, 37075 Göttingen, Germany; (W.W.); (K.I.W.)
- Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Ryan Bowman
- Center for the Neural Basis of Cognition, Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (D.A.K.); (A.R.); (R.B.)
| | - Daniel V. Madison
- Department of Molecular and Cellular Physiology, Stanford University, Palo Alto, CA 94304, USA; (K.D.M.); (D.V.M.)
| | - Katrin I. Willig
- Optical Nanoscopy in Neuroscience, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, 37075 Göttingen, Germany; (W.W.); (K.I.W.)
- Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Alison L. Barth
- Center for the Neural Basis of Cognition, Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA; (D.A.K.); (A.R.); (R.B.)
- Correspondence: ; Tel.: +1-412-268-1198
| |
Collapse
|
22
|
Abstract
The imaging of chromatin, genomic loci, RNAs, and proteins is very important to study their localization, interaction, and coordinated regulation. Recently, several clustered regularly interspaced short palindromic repeats (CRISPR) based imaging methods have been established. The refurbished tool kits utilizing deactivated Cas9 (dCas9) and dCas13 have been established to develop applications of CRISPR-Cas technology beyond genome editing. Here, we review recent advancements in CRISPR-based methods that enable efficient imaging and visualization of chromatin, genomic loci, RNAs, and proteins. RNA aptamers, Pumilio, SuperNova tagging system, molecular beacons, halotag, bimolecular fluorescence complementation, RNA-guided endonuclease in situ labeling, and oligonucleotide-based imaging methods utilizing fluorescent proteins, organic dyes, or quantum dots have been developed to achieve improved fluorescence and signal-to-noise ratio for the imaging of chromatin or genomic loci. RNA-guided RNA targeting CRISPR systems (CRISPR/dCas13) and gene knock-in strategies based on CRISPR/Cas9 mediated site-specific cleavage and DNA repair mechanisms have been employed for efficient RNA and protein imaging, respectively. A few CRISPR-Cas-based methods to investigate the coordinated regulation of DNA-protein, DNA-RNA, or RNA-protein interactions for understanding chromatin dynamics, transcription, and protein function are also available. Overall, the CRISPR-based methods offer a significant improvement in elucidating chromatin organization and dynamics, RNA visualization, and protein imaging. The current and future advancements in CRISPR-based imaging techniques can revolutionize genome biology research for various applications.
Collapse
Affiliation(s)
- Vikram Singh
- School of Computational & Integrative Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Mukesh Jain
- School of Computational & Integrative Sciences, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
23
|
Zhong H, Ceballos CC, Massengill CI, Muniak MA, Ma L, Qin M, Petrie SK, Mao T. High-fidelity, efficient, and reversible labeling of endogenous proteins using CRISPR-based designer exon insertion. eLife 2021; 10:64911. [PMID: 34100715 PMCID: PMC8211447 DOI: 10.7554/elife.64911] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 06/07/2021] [Indexed: 12/26/2022] Open
Abstract
Precise and efficient insertion of large DNA fragments into somatic cells using gene editing technologies to label or modify endogenous proteins remains challenging. Non-specific insertions/deletions (INDELs) resulting from the non-homologous end joining pathway make the process error-prone. Further, the insert is not readily removable. Here, we describe a method called CRISPR-mediated insertion of exon (CRISPIE) that can precisely and reversibly label endogenous proteins using CRISPR/Cas9-based editing. CRISPIE inserts a designer donor module, which consists of an exon encoding the protein sequence flanked by intron sequences, into an intronic location in the target gene. INDELs at the insertion junction will be spliced out, leaving mRNAs nearly error-free. We used CRISPIE to fluorescently label endogenous proteins in mammalian neurons in vivo with previously unachieved efficiency. We demonstrate that this method is broadly applicable, and that the insert can be readily removed later. CRISPIE permits protein sequence insertion with high fidelity, efficiency, and flexibility.
Collapse
Affiliation(s)
- Haining Zhong
- Vollum Institute, Oregon Health & Science University, Portland, United States
| | - Cesar C Ceballos
- Vollum Institute, Oregon Health & Science University, Portland, United States
| | | | - Michael A Muniak
- Vollum Institute, Oregon Health & Science University, Portland, United States
| | - Lei Ma
- Vollum Institute, Oregon Health & Science University, Portland, United States
| | - Maozhen Qin
- Vollum Institute, Oregon Health & Science University, Portland, United States
| | - Stefanie Kaech Petrie
- Department of Neurology, Oregon Health & Science University, Portland, United States
| | - Tianyi Mao
- Vollum Institute, Oregon Health & Science University, Portland, United States
| |
Collapse
|
24
|
Graves AR, Roth RH, Tan HL, Zhu Q, Bygrave AM, Lopez-Ortega E, Hong I, Spiegel AC, Johnson RC, Vogelstein JT, Tward DJ, Miller MI, Huganir RL. Visualizing synaptic plasticity in vivo by large-scale imaging of endogenous AMPA receptors. eLife 2021; 10:66809. [PMID: 34658338 PMCID: PMC8616579 DOI: 10.7554/elife.66809] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 10/16/2021] [Indexed: 02/06/2023] Open
Abstract
Elucidating how synaptic molecules such as AMPA receptors mediate neuronal communication and tracking their dynamic expression during behavior is crucial to understand cognition and disease, but current technological barriers preclude large-scale exploration of molecular dynamics in vivo. We have developed a suite of innovative methodologies that break through these barriers: a new knockin mouse line with fluorescently tagged endogenous AMPA receptors, two-photon imaging of hundreds of thousands of labeled synapses in behaving mice, and computer vision-based automatic synapse detection. Using these tools, we can longitudinally track how the strength of populations of synapses changes during behavior. We used this approach to generate an unprecedentedly detailed spatiotemporal map of synapses undergoing changes in strength following sensory experience. More generally, these tools can be used as an optical probe capable of measuring functional synapse strength across entire brain areas during any behavioral paradigm, describing complex system-wide changes with molecular precision.
Collapse
Affiliation(s)
- Austin R Graves
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States,Kavli Neuroscience Discovery InstituteBaltimoreUnited States
| | - Richard H Roth
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Han L Tan
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Qianwen Zhu
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Alexei M Bygrave
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Elena Lopez-Ortega
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Ingie Hong
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Alina C Spiegel
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States,Kavli Neuroscience Discovery InstituteBaltimoreUnited States
| | - Richard C Johnson
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Joshua T Vogelstein
- Kavli Neuroscience Discovery InstituteBaltimoreUnited States,Center for Imaging Science, Johns Hopkins University School of EngineeringBaltimoreUnited States,Department of Biomedical Engineering, Johns Hopkins UniversityBaltimoreUnited States
| | - Daniel J Tward
- Kavli Neuroscience Discovery InstituteBaltimoreUnited States,Center for Imaging Science, Johns Hopkins University School of EngineeringBaltimoreUnited States,Department of Biomedical Engineering, Johns Hopkins UniversityBaltimoreUnited States
| | - Michael I Miller
- Kavli Neuroscience Discovery InstituteBaltimoreUnited States,Center for Imaging Science, Johns Hopkins University School of EngineeringBaltimoreUnited States,Department of Biomedical Engineering, Johns Hopkins UniversityBaltimoreUnited States
| | - Richard L Huganir
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States,Kavli Neuroscience Discovery InstituteBaltimoreUnited States
| |
Collapse
|
25
|
Nishizono H, Yasuda R, Laviv T. Methodologies and Challenges for CRISPR/Cas9 Mediated Genome Editing of the Mammalian Brain. Front Genome Ed 2020; 2:602970. [PMID: 34713226 PMCID: PMC8525404 DOI: 10.3389/fgeed.2020.602970] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/06/2020] [Indexed: 01/22/2023] Open
Abstract
Neurons and glia are highly polarized cells with extensive subcellular structures extending over large distances from their cell bodies. Previous research has revealed elaborate protein signaling complexes localized within intracellular compartments. Thus, exploring the function and the localization of endogenous proteins is vital to understanding the precise molecular mechanisms underlying the synapse, cellular, and circuit function. Recent advances in CRISPR/Cas9-based genome editing techniques have allowed researchers to rapidly develop transgenic animal models and perform single-cell level genome editing in the mammalian brain. Here, we introduce and comprehensively review the latest techniques for genome-editing in whole animals using fertilized eggs and methods for gene editing in specific neuronal populations in the adult or developing mammalian brain. Finally, we describe the advantages and disadvantages of each technique, as well as the challenges that lie ahead to advance the generation of methodologies for genome editing in the brain using the current CRISPR/Cas9 system.
Collapse
Affiliation(s)
- Hirofumi Nishizono
- Department of Neuronal Signal Transduction, Max Planck Florida Institute for Neuroscience, Jupiter, FL, United States
| | - Ryohei Yasuda
- Department of Neuronal Signal Transduction, Max Planck Florida Institute for Neuroscience, Jupiter, FL, United States
| | | |
Collapse
|
26
|
Verstraelen P, Garcia-Diaz Barriga G, Verschuuren M, Asselbergh B, Nuydens R, Larsen PH, Timmermans JP, De Vos WH. Systematic Quantification of Synapses in Primary Neuronal Culture. iScience 2020; 23:101542. [PMID: 33083769 PMCID: PMC7516133 DOI: 10.1016/j.isci.2020.101542] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 04/30/2020] [Accepted: 09/03/2020] [Indexed: 01/04/2023] Open
Abstract
Most neurological disorders display impaired synaptic connectivity. Hence, modulation of synapse formation may have therapeutic relevance. However, the high density and small size of synapses complicate their quantification. To improve synapse-oriented screens, we analyzed the labeling performance of synapse-targeting antibodies on neuronal cell cultures using segmentation-independent image analysis based on sliding window correlation. When assessing pairwise colocalization, a common readout for mature synapses, overlap was incomplete and confounded by spurious signals. To circumvent this, we implemented a proximity ligation-based approach that only leads to a signal when two markers are sufficiently close. We applied this approach to different marker combinations and demonstrate its utility for detecting synapse density changes in healthy and compromised cultures. Thus, segmentation-independent analysis and exploitation of resident protein proximity increases the sensitivity of synapse quantifications in neuronal cultures and represents a valuable extension to the analytical toolset for in vitro synapse screens.
Collapse
Affiliation(s)
- Peter Verstraelen
- Laboratory of Cell Biology and Histology, University of Antwerp, Wilrijk, Antwerp 2610, Belgium
| | | | - Marlies Verschuuren
- Laboratory of Cell Biology and Histology, University of Antwerp, Wilrijk, Antwerp 2610, Belgium
| | - Bob Asselbergh
- VIB Center for Molecular Neurology, University of Antwerp, Wilrijk, Antwerp 2610, Belgium
| | - Rony Nuydens
- Laboratory of Cell Biology and Histology, University of Antwerp, Wilrijk, Antwerp 2610, Belgium
- Janssen Research and Development, a Division of Janssen Pharmaceutica NV, Beerse, Antwerp 2340, Belgium
| | - Peter H. Larsen
- Janssen Research and Development, a Division of Janssen Pharmaceutica NV, Beerse, Antwerp 2340, Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology and Histology, University of Antwerp, Wilrijk, Antwerp 2610, Belgium
| | - Winnok H. De Vos
- Laboratory of Cell Biology and Histology, University of Antwerp, Wilrijk, Antwerp 2610, Belgium
| |
Collapse
|
27
|
Bensussen S, Shankar S, Ching KH, Zemel D, Ta TL, Mount RA, Shroff SN, Gritton HJ, Fabris P, Vanbenschoten H, Beck C, Man HY, Han X. A Viral Toolbox of Genetically Encoded Fluorescent Synaptic Tags. iScience 2020; 23:101330. [PMID: 32674057 PMCID: PMC7363701 DOI: 10.1016/j.isci.2020.101330] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 04/18/2020] [Accepted: 06/26/2020] [Indexed: 01/16/2023] Open
Abstract
Fibronectin intrabodies generated with mRNA display (FingRs) are a recently developed tool for labeling excitatory or inhibitory synapses, with the benefit of not altering endogenous synaptic protein expression levels or synaptic transmission. Here, we generated a viral vector FingR toolbox that allows for multi-color, neuron-type-specific labeling of excitatory or inhibitory synapses in multiple brain regions. We screened various fluorophores, FingR fusion configurations, and transcriptional control regulations in adeno-associated virus (AAV) and retrovirus vector designs. We report the development of a red FingR variant and demonstrated dual labeling of excitatory and inhibitory synapses in the same cells. Furthermore, we developed cre-inducible FingR AAV variants and demonstrated their utility, finding that the density of inhibitory synapses in aspiny striatal cholinergic interneurons remained unchanged in response to dopamine depletion. Finally, we generated FingR retroviral vectors, which enabled us to track the development of excitatory and inhibitory synapses in hippocampal adult-born granule cells.
Collapse
Affiliation(s)
- Seth Bensussen
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Sneha Shankar
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Kimberley H Ching
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Dana Zemel
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Tina L Ta
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Rebecca A Mount
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Sanaya N Shroff
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Howard J Gritton
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Pierre Fabris
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | | | - Connor Beck
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Heng-Ye Man
- Department of Biology, Boston University, Boston, MA 02215, USA
| | - Xue Han
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
| |
Collapse
|
28
|
Sauerbeck AD, Gangolli M, Reitz SJ, Salyards MH, Kim SH, Hemingway C, Gratuze M, Makkapati T, Kerschensteiner M, Holtzman DM, Brody DL, Kummer TT. SEQUIN Multiscale Imaging of Mammalian Central Synapses Reveals Loss of Synaptic Connectivity Resulting from Diffuse Traumatic Brain Injury. Neuron 2020; 107:257-273.e5. [PMID: 32392471 PMCID: PMC7381374 DOI: 10.1016/j.neuron.2020.04.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 03/04/2020] [Accepted: 04/11/2020] [Indexed: 02/07/2023]
Abstract
The brain's complex microconnectivity underlies its computational abilities and vulnerability to injury and disease. It has been challenging to illuminate the features of this synaptic network due to the small size and dense packing of its elements. Here, we describe a rapid, accessible super-resolution imaging and analysis workflow-SEQUIN-that quantifies central synapses in human tissue and animal models, characterizes their nanostructural and molecular features, and enables volumetric imaging of mesoscale synaptic networks without the production of large histological arrays. Using SEQUIN, we identify cortical synapse loss resulting from diffuse traumatic brain injury, a highly prevalent connectional disorder. Similar synapse loss is observed in three murine models of Alzheimer-related neurodegeneration, where SEQUIN mesoscale mapping identifies regional synaptic vulnerability. These results establish an easily implemented and robust nano-to-mesoscale synapse quantification and characterization method. They furthermore identify a shared mechanism-synaptopathy-between Alzheimer neurodegeneration and its best-established epigenetic risk factor, brain trauma.
Collapse
Affiliation(s)
- Andrew D Sauerbeck
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mihika Gangolli
- McKelvey School of Engineering, Washington University, St. Louis, MO 63130, USA; Currently, Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Sydney J Reitz
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Maverick H Salyards
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Samuel H Kim
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Christopher Hemingway
- Institute of Clinical Neuroimmunology, Ludwig-Maximilians Universität München, Munich 82152, Germany
| | - Maud Gratuze
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tejaswi Makkapati
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Martin Kerschensteiner
- Institute of Clinical Neuroimmunology, Ludwig-Maximilians Universität München, Munich 82152, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich 81377, Germany
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David L Brody
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Currently, Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Terrance T Kummer
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
29
|
BRAIN Initiative: Cutting-Edge Tools and Resources for the Community. J Neurosci 2020; 39:8275-8284. [PMID: 31619497 DOI: 10.1523/jneurosci.1169-19.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 08/09/2019] [Accepted: 08/14/2019] [Indexed: 12/16/2022] Open
Abstract
The overarching goal of the NIH BRAIN (Brain Research through Advancing Innovative Neurotechnologies) Initiative is to advance the understanding of healthy and diseased brain circuit function through technological innovation. Core principles for this goal include the validation and dissemination of the myriad innovative technologies, tools, methods, and resources emerging from BRAIN-funded research. Innovators, BRAIN funding agencies, and non-Federal partners are working together to develop strategies for making these products usable, available, and accessible to the scientific community. Here, we describe several early strategies for supporting the dissemination of BRAIN technologies. We aim to invigorate a dialogue with the neuroscience research and funding community, interdisciplinary collaborators, and trainees about the existing and future opportunities for cultivating groundbreaking research products into mature, integrated, and adaptable research systems. Along with the accompanying Society for Neuroscience 2019 Mini-Symposium, "BRAIN Initiative: Cutting-Edge Tools and Resources for the Community," we spotlight the work of several BRAIN investigator teams who are making progress toward providing tools, technologies, and services for the neuroscience community. These tools access neural circuits at multiple levels of analysis, from subcellular composition to brain-wide network connectivity, including the following: integrated systems for EM- and florescence-based connectomics, advances in immunolabeling capabilities, and resources for recording and analyzing functional connectivity. Investigators describe how the resources they provide to the community will contribute to achieving the goals of the NIH BRAIN Initiative. Finally, in addition to celebrating the contributions of these BRAIN-funded investigators, the Mini-Symposium will illustrate the broader diversity of BRAIN Initiative investments in cutting-edge technologies and resources.
Collapse
|
30
|
Carriba P, Wyatt S, Davies AM. CD40L Reverse Signaling Influences Dendrite Spine Morphology and Expression of PSD-95 and Rho Small GTPases. Front Cell Dev Biol 2020; 8:254. [PMID: 32411702 PMCID: PMC7198883 DOI: 10.3389/fcell.2020.00254] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 03/25/2020] [Indexed: 11/13/2022] Open
Abstract
CD40-activated CD40L reverse signaling is a major physiological regulator of neural process growth from many kinds of developing neurons. Here we have investigated whether CD40L-reverse signaling also influences dendrite spine number and morphology in striatal medium spiny neurons (MSNs). Golgi preparations revealed no differences in the spine density, but because the dendrite arbors of MSNs were larger and branched in Cd40 -/- mice, the total number of spines was greater in Cd40 -/- mice. We also detected more mature spines compared with wild-type littermates. Western blot revealed that MSN cultures from Cd40 -/- mice had significantly less PSD-95 and there were changes in RhoA/B/C and Cdc42. Immunocytochemistry revealed that PSD-95 was clustered in spines in Cd40 -/- neurons compared with more diffuse labeling in Cd40 +/+ neurons. Activation of CD40L-reverse signaling with CD40-Fc prevented the changes observed in Cd40 -/- cultures. Our findings suggest that CD40L-reverse signaling influences dendrite spine morphology and related protein expression and distribution.
Collapse
Affiliation(s)
- Paulina Carriba
- Neuron Development, Neurosciences Department, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Sean Wyatt
- Neuron Development, Neurosciences Department, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Alun M Davies
- Neuron Development, Neurosciences Department, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
31
|
Jiang H, Esparza TJ, Kummer TT, Zhong H, Rettig J, Brody DL. Live Neuron High-Content Screening Reveals Synaptotoxic Activity in Alzheimer Mouse Model Homogenates. Sci Rep 2020; 10:3412. [PMID: 32098978 PMCID: PMC7042280 DOI: 10.1038/s41598-020-60118-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 02/05/2020] [Indexed: 12/28/2022] Open
Abstract
Accurate quantification of synaptic changes is essential for understanding the molecular mechanisms of synaptogenesis, synaptic plasticity, and synaptic toxicity. Here we demonstrate a robust high-content imaging method for the assessment of synaptic changes and apply the method to brain homogenates from an Alzheimer's disease mouse model. Our method uses serial imaging of endogenous fluorescent labeled presynaptic VAMP2 and postsynaptic PSD95 in long-term cultured live primary neurons in 96 well microplates, and uses automatic image analysis to quantify the number of colocalized mature synaptic puncta for the assessment of synaptic changes in live neurons. As a control, we demonstrated that our synaptic puncta assay is at least 10-fold more sensitive to the toxic effects of glutamate than the MTT assay. Using our assay, we have compared synaptotoxic activities in size-exclusion chromatography fractioned protein samples from 3xTg-AD mouse model brain homogenates. Multiple synaptotoxic activities were found in high and low molecular weight fractions. Amyloid-beta immunodepletion alleviated some but not all of the synaptotoxic activities. Although the biochemical entities responsible for the synaptotoxic activities have yet to be determined, these proof-of-concept results demonstrate that this novel assay may have many potential mechanistic and therapeutic applications.
Collapse
Affiliation(s)
- Hao Jiang
- Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, Box 8111, St Louis, Missouri, 63110, USA
| | - Thomas J Esparza
- Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, Box 8111, St Louis, Missouri, 63110, USA
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, 20817, USA
- National Institute of Neurological Disorders and Stroke, 10 Center Drive, Bethesda, Maryland, 20892, USA
| | - Terrance T Kummer
- Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, Box 8111, St Louis, Missouri, 63110, USA
| | - Haining Zhong
- Vollum Institute, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Portland, Oregon, 97239, USA
| | - Jens Rettig
- Department of Physiology, Saarland University, Center for Integrative Physiology and Molecular Medicine (CIPMM), Building 48, Homburg, 66421, Germany
| | - David L Brody
- Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, Box 8111, St Louis, Missouri, 63110, USA.
- National Institute of Neurological Disorders and Stroke, 10 Center Drive, Bethesda, Maryland, 20892, USA.
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, Maryland, 20814, USA.
| |
Collapse
|
32
|
Mikuni T. Genome editing-based approaches for imaging protein localization and dynamics in the mammalian brain. Neurosci Res 2020; 150:2-7. [DOI: 10.1016/j.neures.2019.04.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/15/2019] [Accepted: 04/24/2019] [Indexed: 01/15/2023]
|
33
|
Fluorescence-Based Quantitative Synapse Analysis for Cell Type-Specific Connectomics. eNeuro 2019; 6:ENEURO.0193-19.2019. [PMID: 31548370 PMCID: PMC6873163 DOI: 10.1523/eneuro.0193-19.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/08/2019] [Accepted: 09/11/2019] [Indexed: 12/20/2022] Open
Abstract
Anatomical methods for determining cell type-specific connectivity are essential to inspire and constrain our understanding of neural circuit function. We developed genetically-encoded reagents for fluorescence-synapse labeling and connectivity analysis in brain tissue, using a fluorogen-activating protein (FAP)-coupled or YFP-coupled, postsynaptically-localized neuroligin-1 (NL-1) targeting sequence (FAP/YFPpost). FAPpost expression did not alter mEPSC or mIPSC properties. Sparse AAV-mediated expression of FAP/YFPpost with the cell-filling, red fluorophore dTomato (dTom) enabled high-throughput, compartment-specific detection of putative synapses across diverse neuron types in mouse somatosensory cortex. We took advantage of the bright, far-red emission of FAPpost puncta for multichannel fluorescence alignment of dendrites, FAPpost puncta, and presynaptic neurites in transgenic mice with saturated labeling of parvalbumin (PV), somatostatin (SST), or vasoactive intestinal peptide (VIP)-expressing neurons using Cre-reporter driven expression of YFP. Subtype-specific inhibitory connectivity onto layer 2/3 (L2/3) neocortical pyramidal (Pyr) neurons was assessed using automated puncta detection and neurite apposition. Quantitative and compartment-specific comparisons show that PV inputs are the predominant source of inhibition at both the soma and the dendrites and were particularly concentrated at the primary apical dendrite. SST inputs were interleaved with PV inputs at all secondary-order and higher-order dendritic branches. These fluorescence-based synapse labeling reagents can facilitate large-scale and cell-type specific quantitation of changes in synaptic connectivity across development, learning, and disease states.
Collapse
|
34
|
Green MV, Pengo T, Raybuck JD, Naqvi T, McMullan HM, Hawkinson JE, Marron Fernandez de Velasco E, Muntean BS, Martemyanov KA, Satterfield R, Young SM, Thayer SA. Automated Live-Cell Imaging of Synapses in Rat and Human Neuronal Cultures. Front Cell Neurosci 2019; 13:467. [PMID: 31680875 PMCID: PMC6811609 DOI: 10.3389/fncel.2019.00467] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 10/01/2019] [Indexed: 01/10/2023] Open
Abstract
Synapse loss and dendritic damage correlate with cognitive decline in many neurodegenerative diseases, underlie neurodevelopmental disorders, and are associated with environmental and drug-induced CNS toxicities. However, screening assays designed to measure loss of synaptic connections between live cells are lacking. Here, we describe the design and validation of automated synaptic imaging assay (ASIA), an efficient approach to label, image, and analyze synapses between live neurons. Using viral transduction to express fluorescent proteins that label synapses and an automated computer-controlled microscope, we developed a method to identify agents that regulate synapse number. ASIA is compatible with both confocal and wide-field microscopy; wide-field image acquisition is faster but requires a deconvolution step in the analysis. Both types of images feed into batch processing analysis software that can be run on ImageJ, CellProfiler, and MetaMorph platforms. Primary analysis endpoints are the number of structural synapses and cell viability. Thus, overt cell death is differentiated from subtle changes in synapse density, an important distinction when studying neurodegenerative processes. In rat hippocampal cultures treated for 24 h with 100 μM 2-bromopalmitic acid (2-BP), a compound that prevents clustering of postsynaptic density 95 (PSD95), ASIA reliably detected loss of postsynaptic density 95-enhanced green fluorescent protein (PSD95-eGFP)-labeled synapses in the absence of cell death. In contrast, treatment with 100 μM glutamate produced synapse loss and significant cell death, determined from morphological changes in a binary image created from co-expressed mCherry. Treatment with 3 mM lithium for 24 h significantly increased the number of fluorescent puncta, showing that ASIA also detects synaptogenesis. Proof of concept studies show that cell-specific promoters enable the selective study of inhibitory or principal neurons and that alternative reporter constructs enable quantification of GABAergic or glutamatergic synapses. ASIA can also be used to study synapse loss between human induced pluripotent stem cell (iPSC)-derived cortical neurons. Significant synapse loss in the absence of cell death was detected in the iPSC-derived neuronal cultures treated with either 100 μM 2-BP or 100 μM glutamate for 24 h, while 300 μM glutamate produced synapse loss and cell death. ASIA shows promise for identifying agents that evoke synaptic toxicities and screening for compounds that prevent or reverse synapse loss.
Collapse
Affiliation(s)
- Matthew V. Green
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Thomas Pengo
- Informatics Institute, University of Minnesota, Minneapolis, MN, United States
| | - Jonathan D. Raybuck
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Tahmina Naqvi
- Institute for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, MN, United States
| | - Hannah M. McMullan
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Jon E. Hawkinson
- Institute for Therapeutics Discovery and Development, University of Minnesota, Minneapolis, MN, United States
| | | | - Brian S. Muntean
- Department of Neuroscience, Scripps Research Institute, Jupiter, FL, United States
| | | | - Rachel Satterfield
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, United States
| | - Samuel M. Young
- Department of Anatomy and Cell Biology, Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, United States
- Department of Otolaryngology, University of Iowa, Iowa City, IA, United States
| | - Stanley A. Thayer
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
35
|
Matsuda T, Oinuma I. Imaging endogenous synaptic proteins in primary neurons at single-cell resolution using CRISPR/Cas9. Mol Biol Cell 2019; 30:2838-2855. [PMID: 31509485 PMCID: PMC6789158 DOI: 10.1091/mbc.e19-04-0223] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Fluorescence imaging at single-cell resolution is a crucial approach to analyzing the spatiotemporal regulation of proteins within individual cells of complex neural networks. Here we present a nonviral strategy that enables the tagging of endogenous loci by CRISPR/Cas9-mediated genome editing combined with a nucleofection technique. The method allowed expression of fluorescently tagged proteins at endogenous levels, and we successfully achieved tagging of a presynaptic protein, synaptophysin (Syp), and a postsynaptic protein, PSD-95, in cultured postmitotic neurons. Superresolution fluorescence microscopy of fixed neurons confirmed the identical localization patterns of the tagged proteins to those of endogenous ones verified by immunohistochemistry. The system is also applicable for multiplexed labeling and live-cell imaging. Live imaging with total internal reflection fluorescence microscopy of a single dendritic process of a neuron double-labeled with Syp-mCherry and PSD-95-EGFP revealed the previously undescribed dynamic localization of the proteins synchronously moving along dendritic shafts. Our convenient and versatile strategy is potent for analysis of proteins whose ectopic expressions perturb cellular functions.
Collapse
Affiliation(s)
- Takahiko Matsuda
- Laboratory of Cell and Molecular Biology, Graduate School of Life Science, University of Hyogo, Hyogo 678-1297, Japan
| | - Izumi Oinuma
- Laboratory of Cell and Molecular Biology, Graduate School of Life Science, University of Hyogo, Hyogo 678-1297, Japan.,Institute for Integrated Cell-Material Sciences (iCeMS), Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
36
|
Trivino-Paredes JS, Nahirney PC, Pinar C, Grandes P, Christie BR. Acute slice preparation for electrophysiology increases spine numbers equivalently in the male and female juvenile hippocampus: a DiI labeling study. J Neurophysiol 2019; 122:958-969. [PMID: 31268808 DOI: 10.1152/jn.00332.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hippocampal slices are widely used for in vitro electrophysiological experiments to study underlying mechanisms for synaptic transmission and plasticity, and there is a growing appreciation for sex differences in synaptic plasticity. To date, several studies have shown that the process of making slices from male animals can induce synaptogenesis in cornu ammonis area 1 (CA1) pyramidal cells, but there is a paucity of data for females and other brain regions. In the current study we use microcrystals of the lipophilic carbocyanine dye DiI (1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate) to stain individual neurons in the CA1 and dentate gyrus (DG) hippocampal subfields of postnatal day 21 male and female rats. We show that the preparation of sections for electrophysiology produces significant increases in spines in sections obtained from females, similar to that observed in males. We also show that the procedures used for in vitro electrophysiology also result in significant spine increases in the DG and CA1 subfields. These results demonstrate the utility of this refined DiI procedure for staining neuronal dendrites and spines. They also show, for the first time, that in vitro electrophysiology slice preparations enhance spine numbers on hippocampal cells equivalently in both juvenile females and males.NEW & NOTEWORTHY This study introduces a new DiI technique that elucidates differences in spine numbers in juvenile female and male hippocampus, and shows that slice preparations for hippocampal electrophysiology in vitro may mask these differences.
Collapse
Affiliation(s)
- J S Trivino-Paredes
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - P C Nahirney
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada.,Island Medical Program, University of British Columbia, Victoria, British Columbia, Canada.,Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - C Pinar
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - P Grandes
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada.,Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Vizcaya, Spain.,Achucarro Basque Center for Neuroscience, Science Park of the UPV/EHU, Leioa, Vizcaya, Spain
| | - B R Christie
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada.,Island Medical Program, University of British Columbia, Victoria, British Columbia, Canada.,Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
37
|
Okawa H, Yu WQ, Matti U, Schwarz K, Odermatt B, Zhong H, Tsukamoto Y, Lagnado L, Rieke F, Schmitz F, Wong ROL. Dynamic assembly of ribbon synapses and circuit maintenance in a vertebrate sensory system. Nat Commun 2019; 10:2167. [PMID: 31092821 PMCID: PMC6520400 DOI: 10.1038/s41467-019-10123-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 04/10/2019] [Indexed: 11/11/2022] Open
Abstract
Ribbon synapses transmit information in sensory systems, but their development is not well understood. To test the hypothesis that ribbon assembly stabilizes nascent synapses, we performed simultaneous time-lapse imaging of fluorescently-tagged ribbons in retinal cone bipolar cells (BCs) and postsynaptic densities (PSD95-FP) of retinal ganglion cells (RGCs). Ribbons and PSD95-FP clusters were more stable when these components colocalized at synapses. However, synapse density on ON-alpha RGCs was unchanged in mice lacking ribbons (ribeye knockout). Wildtype BCs make both ribbon-containing and ribbon-free synapses with these GCs even at maturity. Ribbon assembly and cone BC-RGC synapse maintenance are thus regulated independently. Despite the absence of synaptic ribbons, RGCs continued to respond robustly to light stimuli, although quantitative examination of the responses revealed reduced frequency and contrast sensitivity.
Collapse
Affiliation(s)
- Haruhisa Okawa
- Department of Biological Structure, University of Washington, Seattle, 98195, WA, USA
| | - Wan-Qing Yu
- Department of Biological Structure, University of Washington, Seattle, 98195, WA, USA
| | - Ulf Matti
- Department of Neuroanatomy, Medical School Homburg/Saar, Institute for Anatomy and Cell Biology, Saarland University, Homburg/Saar, 66421, Germany
| | - Karin Schwarz
- Department of Neuroanatomy, Medical School Homburg/Saar, Institute for Anatomy and Cell Biology, Saarland University, Homburg/Saar, 66421, Germany
| | | | - Haining Zhong
- Vollum institute, Oregon Health and Science University, Portland, 97239, OR, USA
| | - Yoshihiko Tsukamoto
- Department of Biology, Hyogo College of Medicine, Nishinomiya, 663-8501, Hyogo, Japan
| | - Leon Lagnado
- School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK
| | - Fred Rieke
- Department of Physiology and Biophysics, University of Washington, Seattle, 98195, WA, USA
| | - Frank Schmitz
- Department of Neuroanatomy, Medical School Homburg/Saar, Institute for Anatomy and Cell Biology, Saarland University, Homburg/Saar, 66421, Germany
| | - Rachel O L Wong
- Department of Biological Structure, University of Washington, Seattle, 98195, WA, USA.
| |
Collapse
|
38
|
Verstraelen P, Van Dyck M, Verschuuren M, Kashikar ND, Nuydens R, Timmermans JP, De Vos WH. Image-Based Profiling of Synaptic Connectivity in Primary Neuronal Cell Culture. Front Neurosci 2018; 12:389. [PMID: 29997468 PMCID: PMC6028601 DOI: 10.3389/fnins.2018.00389] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 05/22/2018] [Indexed: 12/04/2022] Open
Abstract
Neurological disorders display a broad spectrum of clinical manifestations. Yet, at the cellular level, virtually all these diseases converge into a common phenotype of dysregulated synaptic connectivity. In dementia, synapse dysfunction precedes neurodegeneration and cognitive impairment by several years, making the synapse a crucial entry point for the development of diagnostic and therapeutic strategies. Whereas high-resolution imaging and biochemical fractionations yield detailed insight into the molecular composition of the synapse, standardized assays are required to quickly gauge synaptic connectivity across large populations of cells under a variety of experimental conditions. Such screening capabilities have now become widely accessible with the advent of high-throughput, high-content microscopy. In this review, we discuss how microscopy-based approaches can be used to extract quantitative information about synaptic connectivity in primary neurons with deep coverage. We elaborate on microscopic readouts that may serve as a proxy for morphofunctional connectivity and we critically analyze their merits and limitations. Finally, we allude to the potential of alternative culture paradigms and integrative approaches to enable comprehensive profiling of synaptic connectivity.
Collapse
Affiliation(s)
- Peter Verstraelen
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Michiel Van Dyck
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Marlies Verschuuren
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | | | - Rony Nuydens
- Janssen Research and Development, Janssen Pharmaceutica N.V., Beerse, Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Winnok H. De Vos
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
- Cell Systems and Imaging, Department of Molecular Biotechnology, Ghent University, Ghent, Belgium
| |
Collapse
|
39
|
Preferential Targeting of Lateral Entorhinal Inputs onto Newly Integrated Granule Cells. J Neurosci 2018; 38:5843-5853. [PMID: 29793975 DOI: 10.1523/jneurosci.1737-17.2018] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 04/26/2018] [Accepted: 05/17/2018] [Indexed: 12/20/2022] Open
Abstract
Mature dentate granule cells in the hippocampus receive input from the entorhinal cortex via the perforant path in precisely arranged lamina, with medial entorhinal axons innervating the middle molecular layer and lateral entorhinal cortex axons innervating the outer molecular layer. Although vastly outnumbered by mature granule cells, adult-generated newborn granule cells play a unique role in hippocampal function, which has largely been attributed to their enhanced excitability and plasticity (Schmidt-Hieber et al., 2004; Ge et al., 2007). Inputs from the medial and lateral entorhinal cortex carry different informational content. Thus, the distribution of inputs onto newly integrated granule cells will affect their function in the circuit. Using retroviral labeling in combination with selective optogenetic activation of medial or lateral entorhinal inputs, we examined the functional innervation and synaptic maturation of newly generated dentate granule cells in the mouse hippocampus. Our results indicate that lateral entorhinal inputs provide the majority of functional innervation of newly integrated granule cells at 21 d postmitosis. Despite preferential functional targeting, the dendritic spine density of immature granule cells was similar in the outer and middle molecular layers, which we speculate could reflect an unequal distribution of shaft synapses. However, chronic blockade of neurotransmitter release of medial entorhinal axons with tetanus toxin disrupted normal synapse development of both medial and lateral entorhinal inputs. Our results support a role for preferential lateral perforant path input onto newly generated neurons in mediating pattern separation, but also indicate that medial perforant path input is necessary for normal synaptic development.SIGNIFICANCE STATEMENT The formation of episodic memories involves the integration of contextual and spatial information. Newly integrated neurons in the dentate gyrus of the hippocampus play a critical role in this process, despite constituting only a minor fraction of the total number of granule cells. Here we demonstrate that these neurons preferentially receive information thought to convey the context of an experience. Each newly integrated granule cell plays this unique role for ∼1 month before reaching maturity.
Collapse
|
40
|
Gantz SC, Ford CP, Morikawa H, Williams JT. The Evolving Understanding of Dopamine Neurons in the Substantia Nigra and Ventral Tegmental Area. Annu Rev Physiol 2018; 80:219-241. [PMID: 28938084 DOI: 10.1146/annurev-physiol-021317-121615] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In recent years, the population of neurons in the ventral tegmental area (VTA) and substantia nigra (SN) has been examined at multiple levels. The results indicate that the projections, neurochemistry, and receptor and ion channel expression in this cell population vary widely. This review centers on the intrinsic properties and synaptic regulation that control the activity of dopamine neurons. Although all dopamine neurons fire action potentials in a pacemaker pattern in the absence of synaptic input, the intrinsic properties that underlie this activity differ considerably. Likewise, the transition into a burst/pause pattern results from combinations of intrinsic ion conductances, inhibitory and excitatory synaptic inputs that differ among this cell population. Finally, synaptic plasticity is a key regulator of the rate and pattern of activity in different groups of dopamine neurons. Through these fundamental properties, the activity of dopamine neurons is regulated and underlies the wide-ranging functions that have been attributed to dopamine.
Collapse
Affiliation(s)
- Stephanie C Gantz
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland 21224, USA
| | - Christopher P Ford
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Hitoshi Morikawa
- Department of Neuroscience and Waggoner Center for Alcohol and Addiction Research, University of Texas, Austin, Texas 78712, USA
| | - John T Williams
- Vollum Institute, Oregon Health Sciences University, Portland, Oregon 97239, USA;
| |
Collapse
|
41
|
Shields BC, Kahuno E, Kim C, Apostolides PF, Brown J, Lindo S, Mensh BD, Dudman JT, Lavis LD, Tadross MR. Deconstructing behavioral neuropharmacology with cellular specificity. Science 2017; 356:356/6333/eaaj2161. [PMID: 28385956 DOI: 10.1126/science.aaj2161] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 02/06/2017] [Indexed: 12/17/2022]
Abstract
Behavior has molecular, cellular, and circuit determinants. However, because many proteins are broadly expressed, their acute manipulation within defined cells has been difficult. Here, we combined the speed and molecular specificity of pharmacology with the cell type specificity of genetic tools. DART (drugs acutely restricted by tethering) is a technique that rapidly localizes drugs to the surface of defined cells, without prior modification of the native target. We first developed an AMPAR antagonist DART, with validation in cultured neuronal assays, in slices of mouse dorsal striatum, and in behaving mice. In parkinsonian animals, motor deficits were causally attributed to AMPARs in indirect spiny projection neurons (iSPNs) and to excess phasic firing of tonically active interneurons (TANs). Together, iSPNs and TANs (i.e., D2 cells) drove akinesia, whereas movement execution deficits reflected the ratio of AMPARs in D2 versus D1 cells. Finally, we designed a muscarinic antagonist DART in one iteration, demonstrating applicability of the method to diverse targets.
Collapse
Affiliation(s)
- Brenda C Shields
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Elizabeth Kahuno
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Charles Kim
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Pierre F Apostolides
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Jennifer Brown
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Sarah Lindo
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Brett D Mensh
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Joshua T Dudman
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Luke D Lavis
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Michael R Tadross
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA. .,Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| |
Collapse
|
42
|
Nolan R, Iliopoulou M, Alvarez L, Padilla-Parra S. Detecting protein aggregation and interaction in live cells: A guide to number and brightness. Methods 2017; 140-141:172-177. [PMID: 29221925 DOI: 10.1016/j.ymeth.2017.12.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 12/01/2017] [Accepted: 12/03/2017] [Indexed: 12/31/2022] Open
Abstract
The possibility to detect and quantify protein-protein interactions with good spatial and temporal resolutions in live cells is crucial in biology. Number and brightness is a powerful approach to detect both protein aggregation/desegregation dynamics and stoichiometry in live cells. Importantly, this technique can be applied in commercial set ups: both camera based and laser scanning microscopes. It provides pixel-by-pixel information on protein oligomeric states. If performed with two colours, the technique can retrieve the stoichiometry of the reaction under study. In this review, we discuss the strengths and weaknesses of the technique, stressing which are the correct acquisition parameters for a given microscope, the main challenges in analysis, and the limitations of the technique.
Collapse
Affiliation(s)
- Rory Nolan
- Wellcome Centre Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Maro Iliopoulou
- Wellcome Centre Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Luis Alvarez
- Wellcome Centre Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Sergi Padilla-Parra
- Wellcome Centre Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Division of Structural Biology, University of Oxford, The Henry Wellcome Building for Genomic Medicine, Headington, Oxford OX3 7BN, UK.
| |
Collapse
|
43
|
Yan Q, Zhai L, Zhang B, Dallman JE. Spatial patterning of excitatory and inhibitory neuropil territories during spinal circuit development. J Comp Neurol 2017; 525:1649-1667. [PMID: 27997694 DOI: 10.1002/cne.24152] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 10/13/2016] [Accepted: 11/14/2016] [Indexed: 01/04/2023]
Abstract
To generate rhythmic motor behaviors, both single neurons and neural circuits require a balance between excitatory inputs that trigger action potentials and inhibitory inputs that promote a stable resting potential (E/I balance). Previous studies have focused on individual neurons and have shown that, over a short spatial scale, excitatory and inhibitory (E/I) synapses tend to form structured territories with inhibitory inputs enriched on cell bodies and proximal dendrites and excitatory inputs on distal dendrites. However, systems-level E/I patterns, at spatial scales larger than single neurons, are largely uncharted. We used immunostaining for PSD-95 and gephyrin postsynaptic scaffolding proteins as proxies for excitatory and inhibitory synapses, respectively, to quantify the numbers and map the distributions of E/I synapses in zebrafish spinal cord at both an embryonic stage and a larval stage. At the embryonic stage, we found that PSD-95 puncta outnumber gephyrin puncta, with the number of gephyrin puncta increasing to match that of PSD-95 puncta at the larval stage. At both stages, PSD-95 puncta are enriched in the most lateral neuropil corresponding to distal dendrites while gephyrin puncta are enriched on neuronal somata and in the medial neuropil. Significantly, similar to synaptic puncta, neuronal processes also exhibit medial-lateral territories at both developmental stages with enrichment of glutamatergic (excitatory) processes laterally and glycinergic (inhibitory) processes medially. This establishment of neuropil excitatory-inhibitory structure largely precedes dendritic arborization of primary motor neurons, suggesting that the structured neuropil could provide a framework for the development of E/I balance at the cellular level. J. Comp. Neurol. 525:1649-1667, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Qing Yan
- Department of Biology, Cox Science Center, University of Miami, 1301 Memorial Drive, Coral Gables, Florida
| | - Lu Zhai
- Department of Biology, Cox Science Center, University of Miami, 1301 Memorial Drive, Coral Gables, Florida
| | - Bo Zhang
- Department of Biology, Cox Science Center, University of Miami, 1301 Memorial Drive, Coral Gables, Florida
| | - Julia E Dallman
- Department of Biology, Cox Science Center, University of Miami, 1301 Memorial Drive, Coral Gables, Florida
| |
Collapse
|
44
|
Yasuda R. Biophysics of Biochemical Signaling in Dendritic Spines: Implications in Synaptic Plasticity. Biophys J 2017; 113:2152-2159. [PMID: 28866426 DOI: 10.1016/j.bpj.2017.07.029] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/12/2017] [Accepted: 07/20/2017] [Indexed: 11/24/2022] Open
Abstract
Dendritic spines are mushroom-shaped postsynaptic compartments that host biochemical signal cascades important for synaptic plasticity and, ultimately, learning and memory. Signaling events in spines involve a signaling network composed of hundreds of signaling proteins interacting with each other extensively. Synaptic plasticity is typically induced by Ca2+ elevation in spines, which activates a variety of signaling pathways. This leads to changes in the actin cytoskeleton and membrane dynamics, which in turn causes structural and functional changes of the spine. Recent studies have demonstrated that the activities of these proteins have a variety of spatiotemporal patterns, which orchestrate signaling activity in different subcellular compartments at different timescales. The diffusion and the decay kinetics of signaling molecules play important roles in determining the degree of their spatial spreading, and thereby the degree of the spine specificity of the signaling pathway.
Collapse
Affiliation(s)
- Ryohei Yasuda
- Max Planck Florida Institute for Neuroscience, Jupiter, Florida.
| |
Collapse
|
45
|
Funk AJ, Mielnik CA, Koene R, Newburn E, Ramsey AJ, Lipska BK, McCullumsmith RE. Postsynaptic Density-95 Isoform Abnormalities in Schizophrenia. Schizophr Bull 2017; 43:891-899. [PMID: 28126896 PMCID: PMC5472126 DOI: 10.1093/schbul/sbw173] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
BACKGROUND Postsynaptic density-95 (PSD-95) protein expression is dysregulated in schizophrenia in a variety of brain regions. We have designed experiments to examine PSD-95 mRNA splice variant expression in the dorsolateral prefrontal cortex from subjects with schizophrenia. METHODS We performed quantitative PCR and western blot analysis to measure PSD-95 expression in schizophrenia vs control subjects, rodent haloperidol treatment studies, rodent postmortem interval studies, and GluN1 knockdown (KD) mice vs controls. RESULTS We found decreased mRNA expression of beta (t = 4.506, df = 383, P < .0001) and truncated (t = 3.378, df = 383, P = .0008) isoforms of PSD-95, whereas alpha was unchanged. Additionally, we found decreased PSD-95 protein expression in schizophrenia (t = 2.746, df = 71, P = .0076). We found no correlation between PSD-95 protein and alpha, beta, or truncated mRNA isoforms in schizophrenia. PSD-95 beta transcript was increased (t = 3.346, df = 14, P < .05) in the GluN1 KD mouse model of schizophrenia. There was an increase in PSD-95 alpha mRNA expression (t = 2.905, df = 16, P < .05) in rats following long-term haloperidol administration. CONCLUSIONS Our findings describe a unique pathophysiology of specific PSD-95 isoform dysregulation in schizophrenia, chronic neuroleptic treatment, and a genetic lesion mouse model of drastically reduced N-methyl-d-aspartate receptor (NMDAR) complex expression. These data indicate that regulation of PSD-95 is multifaceted, may be isoform specific, and biologically relevant for synaptic signaling function. Specifically, NMDAR-mediated synaptic remodeling, and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor trafficking and interaction may be impaired in schizophrenia by decreased PSD-95 beta and truncated expression (respectively). Further, increased PSD-95 beta transcript in the GluN1 KD mouse model poses a potential compensatory rescue of NMDAR-mediated function via increased postsynaptic throughput of the severely reduced GluN1 signal. Together, these data propose that disruption of excitatory signaling complexes through genetic (GluN1 KD), pharmacologic (antipsychotics), or disease (schizophrenia) mechanisms specifically dysregulates PSD-95 expression.
Collapse
Affiliation(s)
- Adam J. Funk
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Catharine A. Mielnik
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Rachael Koene
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH
| | | | - Amy J. Ramsey
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Barbara K. Lipska
- Human Brain Collection Core, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, MD,Co-senior authors
| | - Robert E. McCullumsmith
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati College of Medicine, Cincinnati, OH;,Co-senior authors
| |
Collapse
|
46
|
Protein Crowding within the Postsynaptic Density Can Impede the Escape of Membrane Proteins. J Neurosci 2016; 36:4276-95. [PMID: 27076425 DOI: 10.1523/jneurosci.3154-15.2016] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 02/19/2016] [Indexed: 01/03/2023] Open
Abstract
UNLABELLED Mechanisms regulating lateral diffusion and positioning of glutamate receptors within the postsynaptic density (PSD) determine excitatory synaptic strength. Scaffold proteins in the PSD are abundant receptor binding partners, yet electron microscopy suggests that the PSD is highly crowded, potentially restricting the diffusion of receptors regardless of binding. However, the contribution of macromolecular crowding to receptor retention remains poorly understood. We combined experimental and computational approaches to test the effect of synaptic crowding on receptor movement and positioning in Sprague Dawley rat hippocampal neurons. We modeled AMPA receptor diffusion in synapses where the distribution of scaffold proteins was determined from photoactivated localization microscopy experiments, and receptor-scaffold association and dissociation rates were adjusted to fit single-molecule tracking and fluorescence recovery measurements. Simulations predicted that variation of receptor size strongly influences the fractional synaptic area the receptor may traverse, and the proportion that may exchange in and out of the synapse. To test the model experimentally, we designed a set of novel transmembrane (TM) probes. A single-pass TM protein with one PDZ binding motif concentrated in the synapse as do AMPARs yet was more mobile there than the much larger AMPAR. Furthermore, either the single binding motif or an increase in cytoplasmic bulk through addition of a single GFP slowed synaptic movement of a small TM protein. These results suggest that both crowding and binding limit escape of AMPARs from the synapse. Moreover, tight protein packing within the PSD may modulate the synaptic dwell time of many TM proteins important for synaptic function. SIGNIFICANCE STATEMENT Small alterations to the distribution within synapses of key transmembrane proteins, such as receptors, can dramatically change synaptic strength. Indeed, many diseases are thought to unbalance neural circuit function in this manner. Processes that regulate this in healthy synapses are unclear, however. By combining computer simulations with imaging methods that examined protein dynamics at multiple scales in space and time, we showed that both steric effects and protein-protein binding each regulate the mobility of receptors in the synapse. Our findings extend our knowledge of the synapse as a crowded environment that counteracts molecular diffusion, and support the idea that both molecular collisions and biochemical binding can be involved in the regulation of neural circuit performance.
Collapse
|
47
|
Lee H, Oh WC, Seong J, Kim J. Advanced Fluorescence Protein-Based Synapse-Detectors. Front Synaptic Neurosci 2016; 8:16. [PMID: 27445785 PMCID: PMC4927625 DOI: 10.3389/fnsyn.2016.00016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 06/13/2016] [Indexed: 11/13/2022] Open
Abstract
The complex information-processing capabilities of the central nervous system emerge from intricate patterns of synaptic input-output relationships among various neuronal circuit components. Understanding these capabilities thus requires a precise description of the individual synapses that comprise neural networks. Recent advances in fluorescent protein engineering, along with developments in light-favoring tissue clearing and optical imaging techniques, have rendered light microscopy (LM) a potent candidate for large-scale analyses of synapses, their properties, and their connectivity. Optically imaging newly engineered fluorescent proteins (FPs) tagged to synaptic proteins or microstructures enables the efficient, fine-resolution illumination of synaptic anatomy and function in large neural circuits. Here we review the latest progress in fluorescent protein-based molecular tools for imaging individual synapses and synaptic connectivity. We also identify associated technologies in gene delivery, tissue processing, and computational image analysis that will play a crucial role in bridging the gap between synapse- and system-level neuroscience.
Collapse
Affiliation(s)
- Hojin Lee
- Center for Functional Connectomics, Korea Institute of Science and TechnologySeoul, South Korea; Neuroscience Program, Korea University of Science and TechnologyDaejeon, South Korea
| | - Won Chan Oh
- Center for Functional Connectomics, Korea Institute of Science and Technology Seoul, South Korea
| | - Jihye Seong
- Neuroscience Program, Korea University of Science and TechnologyDaejeon, South Korea; Center for Diagnosis Treatment Care of Dementia, Korea Institute of Science and TechnologySeoul, South Korea
| | - Jinhyun Kim
- Center for Functional Connectomics, Korea Institute of Science and TechnologySeoul, South Korea; Neuroscience Program, Korea University of Science and TechnologyDaejeon, South Korea
| |
Collapse
|
48
|
Molecular evidence of synaptic pathology in the CA1 region in schizophrenia. NPJ SCHIZOPHRENIA 2016; 2:16022. [PMID: 27430010 PMCID: PMC4944906 DOI: 10.1038/npjschz.2016.22] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Revised: 05/21/2016] [Accepted: 05/23/2016] [Indexed: 11/09/2022]
Abstract
Alterations of postsynaptic density (PSD)95-complex proteins in schizophrenia ostensibly induce deficits in synaptic plasticity, the molecular process underlying cognitive functions. Although some PSD95-complex proteins have been previously examined in the hippocampus in schizophrenia, the status of other equally important molecules is unclear. This is especially true in the cornu ammonis (CA)1 hippocampal subfield, a region that is critically involved in the pathophysiology of the illness. We thus performed a quantitative immunoblot experiment to examine PSD95 and several of its associated proteins in the CA1 region, using post mortem brain samples derived from schizophrenia subjects with age-, sex-, and post mortem interval-matched controls (n=20/group). Our results indicate a substantial reduction in PSD95 protein expression (-61.8%). Further analysis showed additional alterations to the scaffold protein Homer1 (Homer1a: +42.9%, Homer1b/c: -24.6%), with a twofold reduction in the ratio of Homer1b/c:Homer1a isoforms (P=0.011). Metabotropic glutamate receptor 1 (mGluR1) protein levels were significantly reduced (-32.7%), and Preso, a protein that supports interactions between Homer1 or PSD95 with mGluR1, was elevated (+83.3%). Significant reduction in synaptophysin (-27.8%) was also detected, which is a validated marker of synaptic density. These findings support the presence of extensive molecular abnormalities to PSD95 and several of its associated proteins in the CA1 region in schizophrenia, offering a small but significant step toward understanding how proteins in the PSD are altered in the schizophrenia brain, and their relevance to overall hippocampal and cognitive dysfunction in the illness.
Collapse
|
49
|
Mikuni T, Nishiyama J, Sun Y, Kamasawa N, Yasuda R. High-Throughput, High-Resolution Mapping of Protein Localization in Mammalian Brain by In Vivo Genome Editing. Cell 2016; 165:1803-1817. [PMID: 27180908 DOI: 10.1016/j.cell.2016.04.044] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/14/2016] [Accepted: 04/13/2016] [Indexed: 12/25/2022]
Abstract
A scalable and high-throughput method to identify precise subcellular localization of endogenous proteins is essential for integrative understanding of a cell at the molecular level. Here, we developed a simple and generalizable technique to image endogenous proteins with high specificity, resolution, and contrast in single cells in mammalian brain tissue. The technique, single-cell labeling of endogenous proteins by clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9-mediated homology-directed repair (SLENDR), uses in vivo genome editing to insert a sequence encoding an epitope tag or a fluorescent protein to a gene of interest by CRISPR-Cas9-mediated homology-directed repair (HDR). Single-cell, HDR-mediated genome editing was achieved by delivering the editing machinery to dividing neuronal progenitors through in utero electroporation. We demonstrate that SLENDR allows rapid determination of the localization and dynamics of many endogenous proteins in various cell types, regions, and ages in the brain. Thus, SLENDR provides a high-throughput platform to map the subcellular localization of endogenous proteins with the resolution of micro- to nanometers in the brain.
Collapse
Affiliation(s)
- Takayasu Mikuni
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Jun Nishiyama
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA.
| | - Ye Sun
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA; Integrative Program in Biology and Neuroscience, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Naomi Kamasawa
- Electron Microscopy Core Facility, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Ryohei Yasuda
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA.
| |
Collapse
|
50
|
Hage TA, Sun Y, Khaliq ZM. Electrical and Ca(2+) signaling in dendritic spines of substantia nigra dopaminergic neurons. eLife 2016; 5. [PMID: 27163179 PMCID: PMC4900803 DOI: 10.7554/elife.13905] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 05/09/2016] [Indexed: 11/13/2022] Open
Abstract
Little is known about the density and function of dendritic spines on midbrain dopamine neurons, or the relative contribution of spine and shaft synapses to excitability. Using Ca(2+) imaging, glutamate uncaging, fluorescence recovery after photobleaching and transgenic mice expressing labeled PSD-95, we comparatively analyzed electrical and Ca(2+) signaling in spines and shaft synapses of dopamine neurons. Dendritic spines were present on dopaminergic neurons at low densities in live and fixed tissue. Uncaging-evoked potential amplitudes correlated inversely with spine length but positively with the presence of PSD-95. Spine Ca(2+) signals were less sensitive to hyperpolarization than shaft synapses, suggesting amplification of spine head voltages. Lastly, activating spines during pacemaking, we observed an unexpected enhancement of spine Ca(2+) midway throughout the spike cycle, likely involving recruitment of NMDA receptors and voltage-gated conductances. These results demonstrate functionality of spines in dopamine neurons and reveal a novel modulation of spine Ca(2+) signaling during pacemaking.
Collapse
Affiliation(s)
- Travis A Hage
- Cellular Neurophysiology Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, United States
| | - Yujie Sun
- Cellular Neurophysiology Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, United States
| | - Zayd M Khaliq
- Cellular Neurophysiology Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, United States
| |
Collapse
|