251
|
El Amki M, Glück C, Binder N, Middleham W, Wyss MT, Weiss T, Meister H, Luft A, Weller M, Weber B, Wegener S. Neutrophils Obstructing Brain Capillaries Are a Major Cause of No-Reflow in Ischemic Stroke. Cell Rep 2021; 33:108260. [PMID: 33053341 DOI: 10.1016/j.celrep.2020.108260] [Citation(s) in RCA: 147] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 08/18/2020] [Accepted: 09/21/2020] [Indexed: 12/29/2022] Open
Abstract
Despite successful clot retrieval in large vessel occlusion stroke, ∼50% of patients have an unfavorable clinical outcome. The mechanisms underlying this functional reperfusion failure remain unknown, and therapeutic options are lacking. In the thrombin-model of middle cerebral artery (MCA) stroke in mice, we show that, despite successful thrombolytic recanalization of the proximal MCA, cortical blood flow does not fully recover. Using in vivo two-photon imaging, we demonstrate that this is due to microvascular obstruction of ∼20%-30% of capillaries in the infarct core and penumbra by neutrophils adhering to distal capillary segments. Depletion of circulating neutrophils using an anti-Ly6G antibody restores microvascular perfusion without increasing the rate of hemorrhagic complications. Strikingly, infarct size and functional deficits are smaller in mice treated with anti-Ly6G. Thus, we propose neutrophil stalling of brain capillaries to contribute to reperfusion failure, which offers promising therapeutic avenues for ischemic stroke.
Collapse
Affiliation(s)
- Mohamad El Amki
- Department of Neurology, University Hospital and University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - Chaim Glück
- Experimental Imaging and Neuroenergetics, Institute of Pharmacology and Toxicology, University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - Nadine Binder
- Department of Neurology, University Hospital and University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - William Middleham
- Department of Neurology, University Hospital and University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - Matthias T Wyss
- Experimental Imaging and Neuroenergetics, Institute of Pharmacology and Toxicology, University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - Tobias Weiss
- Department of Neurology, University Hospital and University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - Hanna Meister
- Department of Neurology, University Hospital and University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - Andreas Luft
- Department of Neurology, University Hospital and University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - Bruno Weber
- Experimental Imaging and Neuroenergetics, Institute of Pharmacology and Toxicology, University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland.
| | - Susanne Wegener
- Department of Neurology, University Hospital and University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland.
| |
Collapse
|
252
|
Arttamangkul S, Platt EJ, Carroll J, Farrens D. Functional independence of endogenous µ- and δ-opioid receptors co-expressed in cholinergic interneurons. eLife 2021; 10:69740. [PMID: 34477106 PMCID: PMC8718112 DOI: 10.7554/elife.69740] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 08/31/2021] [Indexed: 12/18/2022] Open
Abstract
Class A G-protein-coupled receptors (GPCRs) normally function as monomers, although evidence from heterologous expression systems suggests that they may sometimes form homodimers and/or heterodimers. This study aims to evaluate possible functional interplay of endogenous µ- and δ-opioid receptors (MORs and DORs) in mouse neurons. Detecting GPCR dimers in native tissues, however, has been challenging. Previously, MORs and DORs co-expressed in transfected cells have been reported to form heterodimers, and their possible co-localization in neurons has been studied in knock-in mice expressing genetically engineered receptors fused to fluorescent proteins. Here, we find that single cholinergic neurons in the mouse striatum endogenously express both MORs and DORs. The receptors on neurons from live brain slices were fluorescently labeled with a ligand-directed labeling reagent, NAI-A594. The selective activation of MORs and DORs, with DAMGO (µ-agonist) and deltorphin (δ-agonist) inhibited spontaneous firing in all cells examined. In the continued presence of agonist, the firing rate returned to baseline as the result of receptor desensitization with the application of deltorphin but was less observed with the application of DAMGO. In addition, agonist-induced internalization of DORs but not MORs was detected. When MORs and DORs were activated simultaneously with [Met5]-enkephalin, desensitization of MORs was facilitated but internalization was not increased. Together, these results indicate that while MORs and DORs are expressed in single striatal cholinergic interneurons, the two receptors function independently.
Collapse
Affiliation(s)
| | - Emily J Platt
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, United States
| | - James Carroll
- Surgery, Oregon Health and Science University, Portland, United States
| | - David Farrens
- Department of Biochemistry and Molecular Biology, School of Medicine, Oregon Health and Science University, Portland, United States
| |
Collapse
|
253
|
Climer JR, Dombeck DA. Information Theoretic Approaches to Deciphering the Neural Code with Functional Fluorescence Imaging. eNeuro 2021; 8:ENEURO.0266-21.2021. [PMID: 34433574 PMCID: PMC8474651 DOI: 10.1523/eneuro.0266-21.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/22/2021] [Accepted: 08/04/2021] [Indexed: 11/21/2022] Open
Abstract
Information theoretic metrics have proven useful in quantifying the relationship between behaviorally relevant parameters and neuronal activity with relatively few assumptions. However, these metrics are typically applied to action potential (AP) recordings and were not designed for the slow timescales and variable amplitudes typical of functional fluorescence recordings (e.g., calcium imaging). The lack of research guidelines on how to apply and interpret these metrics with fluorescence traces means the neuroscience community has yet to realize the power of information theoretic metrics. Here, we used computational methods to create mock AP traces with known amounts of information. From these, we generated fluorescence traces and examined the ability of different information metrics to recover the known information values. We provide guidelines for how to use information metrics when applying them to functional fluorescence and demonstrate their appropriate application to GCaMP6f population recordings from mouse hippocampal neurons imaged during virtual navigation.
Collapse
Affiliation(s)
- Jason R Climer
- Department of Neurobiology, Northwestern University, Evanston, 60208 IL
| | - Daniel A Dombeck
- Department of Neurobiology, Northwestern University, Evanston, 60208 IL
| |
Collapse
|
254
|
Rupprecht P, Carta S, Hoffmann A, Echizen M, Blot A, Kwan AC, Dan Y, Hofer SB, Kitamura K, Helmchen F, Friedrich RW. A database and deep learning toolbox for noise-optimized, generalized spike inference from calcium imaging. Nat Neurosci 2021; 24:1324-1337. [PMID: 34341584 PMCID: PMC7611618 DOI: 10.1038/s41593-021-00895-5] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 06/23/2021] [Indexed: 02/06/2023]
Abstract
Inference of action potentials ('spikes') from neuronal calcium signals is complicated by the scarcity of simultaneous measurements of action potentials and calcium signals ('ground truth'). In this study, we compiled a large, diverse ground truth database from publicly available and newly performed recordings in zebrafish and mice covering a broad range of calcium indicators, cell types and signal-to-noise ratios, comprising a total of more than 35 recording hours from 298 neurons. We developed an algorithm for spike inference (termed CASCADE) that is based on supervised deep networks, takes advantage of the ground truth database, infers absolute spike rates and outperforms existing model-based algorithms. To optimize performance for unseen imaging data, CASCADE retrains itself by resampling ground truth data to match the respective sampling rate and noise level; therefore, no parameters need to be adjusted by the user. In addition, we developed systematic performance assessments for unseen data, openly released a resource toolbox and provide a user-friendly cloud-based implementation.
Collapse
Affiliation(s)
- Peter Rupprecht
- Brain Research Institute, University of Zürich, Zurich, Switzerland.
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland.
| | - Stefano Carta
- Brain Research Institute, University of Zürich, Zurich, Switzerland
| | - Adrian Hoffmann
- Brain Research Institute, University of Zürich, Zurich, Switzerland
| | - Mayumi Echizen
- Department of Neurophysiology, University of Tokyo, Tokyo, Japan
- Department of Anesthesiology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Antonin Blot
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, London, United Kingdom
- Biozentrum, University of Basel, Basel, Switzerland
| | - Alex C Kwan
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Yang Dan
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley CA, USA
| | - Sonja B Hofer
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, London, United Kingdom
- Biozentrum, University of Basel, Basel, Switzerland
| | - Kazuo Kitamura
- Department of Neurophysiology, University of Tokyo, Tokyo, Japan
- Department of Neurophysiology, University of Yamanashi, Yamanashi, Japan
| | - Fritjof Helmchen
- Brain Research Institute, University of Zürich, Zurich, Switzerland.
| | - Rainer W Friedrich
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland.
- University of Basel, Basel, Switzerland.
| |
Collapse
|
255
|
Schweigmann M, Caudal LC, Stopper G, Scheller A, Koch KP, Kirchhoff F. Versatile Surface Electrodes for Combined Electrophysiology and Two-Photon Imaging of the Mouse Central Nervous System. Front Cell Neurosci 2021; 15:720675. [PMID: 34447299 PMCID: PMC8383317 DOI: 10.3389/fncel.2021.720675] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/19/2021] [Indexed: 11/23/2022] Open
Abstract
Understanding and modulating CNS function in physiological as well as pathophysiological contexts remains a significant ambition in research and clinical applications. The investigation of the multifaceted CNS cell types including their interactions and contributions to neural function requires a combination of the state-of-the-art in vivo electrophysiology and imaging techniques. We developed a novel type of liquid crystal polymer (LCP) surface micro-electrode manufactured in three customized designs with up to 16 channels for recording and stimulation of brain activity. All designs include spare central spaces for simultaneous 2P-imaging. Nanoporous platinum-plated contact sites ensure a low impedance and high current transfer. The epidural implantation of the LCP micro-electrodes could be combined with standard cranial window surgery. The epidurally positioned electrodes did not only display long-term biocompatibility, but we also observed an additional stabilization of the underlying CNS tissue. We demonstrate the electrode’s versatility in combination with in vivo 2P-imaging by monitoring anesthesia-awake cycles of transgenic mice with GCaMP3 expression in neurons or astrocytes. Cortical stimulation and simultaneous 2P Ca2+ imaging in neurons or astrocytes highlighted the astrocytes’ integrative character in neuronal activity processing. Furthermore, we confirmed that spontaneous astroglial Ca2+ signals are dampened under anesthesia, while evoked signals in neurons and astrocytes showed stronger dependency on stimulation intensity rather than on various levels of anesthesia. Finally, we show that the electrodes provide recordings of the electrocorticogram (ECoG) with a high signal-to noise ratio and spatial signal differences which help to decipher brain activity states during experimental procedures. Summarizing, the novel LCP surface micro-electrode is a versatile, convenient, and reliable tool to investigate brain function in vivo.
Collapse
Affiliation(s)
- Michael Schweigmann
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany.,Department of Electrical Engineering, Trier University of Applied Sciences, Trier, Germany
| | - Laura C Caudal
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Gebhard Stopper
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Anja Scheller
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Klaus P Koch
- Department of Electrical Engineering, Trier University of Applied Sciences, Trier, Germany
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| |
Collapse
|
256
|
Shimizu T, Murakoshi H, Matsumoto H, Ichino K, Hattori A, Ueno S, Ishida A, Tajiri N, Hida H. Tension Sensor Based on Fluorescence Resonance Energy Transfer Reveals Fiber Diameter-Dependent Mechanical Factors During Myelination. Front Cell Neurosci 2021; 15:685044. [PMID: 34408628 PMCID: PMC8364977 DOI: 10.3389/fncel.2021.685044] [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: 03/24/2021] [Accepted: 07/13/2021] [Indexed: 11/15/2022] Open
Abstract
Oligodendrocytes (OLs) form a myelin sheath around neuronal axons to increase conduction velocity of action potential. Although both large and small diameter axons are intermingled in the central nervous system (CNS), the number of myelin wrapping is related to the axon diameter, such that the ratio of the diameter of the axon to that of the entire myelinated-axon unit is optimal for each axon, which is required for exerting higher brain functions. This indicates there are unknown axon diameter-dependent factors that control myelination. We tried to investigate physical factors to clarify the mechanisms underlying axon diameter-dependent myelination. To visualize OL-generating forces during myelination, a tension sensor based on fluorescence resonance energy transfer (FRET) was used. Polystyrene nanofibers with varying diameters similar to neuronal axons were prepared to investigate biophysical factors regulating the OL-axon interactions. We found that higher tension was generated at OL processes contacting larger diameter fibers compared with smaller diameter fibers. Additionally, OLs formed longer focal adhesions (FAs) on larger diameter axons and shorter FAs on smaller diameter axons. These results suggest that OLs respond to the fiber diameter and activate mechanotransduction initiated at FAs, which controls their cytoskeletal organization and myelin formation. This study leads to the novel and interesting idea that physical factors are involved in myelin formation in response to axon diameter.
Collapse
Affiliation(s)
- Takeshi Shimizu
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hideji Murakoshi
- Supportive Center for Brain Research, National Institute for Physiological Sciences, Okazaki, Japan.,Department of Physiological Sciences, The Graduate University for Advanced Studies, Okazaki, Japan
| | - Hidetoshi Matsumoto
- Department of Materials Science and Engineering, Tokyo Institute of Technology, Meguro, Japan
| | - Kota Ichino
- Department of Materials Science and Engineering, Tokyo Institute of Technology, Meguro, Japan
| | - Atsunori Hattori
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shinya Ueno
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Akimasa Ishida
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Naoki Tajiri
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hideki Hida
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
257
|
Müller FE, Cherkas V, Stopper G, Caudal LC, Stopper L, Kirchhoff F, Henneberger C, Ponimaskin EG, Zeug A. Elucidating regulators of astrocytic Ca 2+ signaling via multi-threshold event detection (MTED). Glia 2021; 69:2798-2811. [PMID: 34388285 DOI: 10.1002/glia.24070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 01/23/2023]
Abstract
Recent achievements in indicator optimization and imaging techniques promote the advancement of functional imaging to decipher complex signaling processes in living cells, such as Ca2+ activity patterns. Astrocytes are important regulators of the brain network and well known for their highly complex morphology and spontaneous Ca2+ activity. However, the astrocyte community is lacking standardized methods to analyze and interpret Ca2+ activity recordings, hindering global comparisons. Here, we present a biophysically-based analytical concept for deciphering the complex spatio-temporal changes of Ca2+ biosensor fluorescence for understanding the underlying signaling mechanisms. We developed a pixel-based multi-threshold event detection (MTED) analysis of multidimensional data, which accounts for signal strength as an additional signaling dimension and provides the experimenter with a comprehensive toolbox for a differentiated and in-depth characterization of fluorescence signals. MTED was validated by analyzing astrocytic Ca2+ activity across Ca2+ indicators, imaging setups, and model systems from primary cell culture to awake, head-fixed mice. We identified extended Ca2+ activity at 25°C compared to 37°C physiological body temperature and dissected how neuronal activity shapes long-lasting astrocytic Ca2+ activity. Our MTED strategy, as a parameter-free approach, is easily transferrable to other fluorescent indicators and biosensors and embraces the additional dimensionality of signaling activity strength. It will also advance the definition of standardized procedures and parameters to improve comparability of research data and reports.
Collapse
Affiliation(s)
| | - Volodymyr Cherkas
- Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Gebhard Stopper
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Laura C Caudal
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Laura Stopper
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Frank Kirchhoff
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Christian Henneberger
- Institute of Cellular Neurosciences, Medical Faculty, University of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Institute of Neurology, University College London, London, UK
| | | | - Andre Zeug
- Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
258
|
Carretero-Rodriguez L, Guðjónsdóttir R, Poparic I, Reilly ML, Chol M, Bianco IH, Chiapello M, Feret R, Deery MJ, Guthrie S. The Rac-GAP alpha2-Chimaerin Signals via CRMP2 and Stathmins in the Development of the Ocular Motor System. J Neurosci 2021; 41:6652-6672. [PMID: 34168008 PMCID: PMC8336708 DOI: 10.1523/jneurosci.0983-19.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 04/02/2021] [Accepted: 04/05/2021] [Indexed: 11/21/2022] Open
Abstract
A precise sequence of axon guidance events is required for the development of the ocular motor system. Three cranial nerves grow toward, and connect with, six extraocular muscles in a stereotyped pattern, to control eye movements. The signaling protein alpha2-chimaerin (α2-CHN) plays a pivotal role in the formation of the ocular motor system; mutations in CHN1, encoding α2-CHN, cause the human eye movement disorder Duane Retraction Syndrome (DRS). Our research has demonstrated that the manipulation of α2-chn signaling in the zebrafish embryo leads to ocular motor axon wiring defects, although the signaling cascades regulated by α2-chn remain poorly understood. Here, we demonstrate that several cytoskeletal regulatory proteins-collapsin response mediator protein 2 (CRMP2; encoded by the gene dpysl2), stathmin1, and stathmin 2-bind to α2-CHN. dpysl2, stathmin1, and especially stathmin2 are expressed by ocular motor neurons. We find that the manipulation of dpysl2 and of stathmins in zebrafish larvae leads to defects in both the axon wiring of the ocular motor system and the optokinetic reflex, impairing horizontal eye movements. Knockdowns of these molecules in zebrafish larvae of either sex caused axon guidance phenotypes that included defasciculation and ectopic branching; in some cases, these phenotypes were reminiscent of DRS. chn1 knock-down phenotypes were rescued by the overexpression of CRMP2 and STMN1, suggesting that these proteins act in the same signaling pathway. These findings suggest that CRMP2 and stathmins signal downstream of α2-CHN to orchestrate ocular motor axon guidance and to control eye movements.SIGNIFICANCE STATEMENT The precise control of eye movements is crucial for the life of vertebrate animals, including humans. In humans, this control depends on the arrangement of nerve wiring of the ocular motor system, composed of three nerves and six muscles, a system that is conserved across vertebrate phyla. Mutations in the protein alpha2-chimaerin have previously been shown to cause eye movement disorders (squint) and axon wiring defects in humans. Our recent work has unraveled how alpha2-chimaerin coordinates axon guidance of the ocular motor system in animal models. In this article, we demonstrate key roles for the proteins CRMP2 and stathmin 1/2 in the signaling pathway orchestrated by alpha2-chimaerin, potentially giving insight into the etiology of eye movement disorders in humans.
Collapse
Affiliation(s)
| | | | - Ivana Poparic
- Department of Developmental Neurobiology, King's College London, London SE1 1UL, United Kingdom
| | | | - Mary Chol
- Department of Developmental Neurobiology, King's College London, London SE1 1UL, United Kingdom
| | - Isaac H Bianco
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, United Kingdom
| | - Marco Chiapello
- Cambridge Centre for Proteomics, Cambridge Systems Biology Centre, Department of Biochemistry, University of Cambridge, Cambridge CB2 1QR, United Kingdom
| | - Renata Feret
- Institute for Sustainable Plant Protection, National Research Council, 10135 Torino, Italy
| | - Michael J Deery
- Institute for Sustainable Plant Protection, National Research Council, 10135 Torino, Italy
| | - Sarah Guthrie
- School of Life Sciences, University of Sussex, Brighton BN7 9QG, United Kingdom
| |
Collapse
|
259
|
Condon AF, Robinson BG, Asad N, Dore TM, Tian L, Williams JT. The residence of synaptically released dopamine on D2 autoreceptors. Cell Rep 2021; 36:109465. [PMID: 34348146 PMCID: PMC8351352 DOI: 10.1016/j.celrep.2021.109465] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 05/24/2021] [Accepted: 07/08/2021] [Indexed: 11/30/2022] Open
Abstract
Neuromodulation mediated by synaptically released endogenous transmitters acting in G-protein-coupled receptors (GPCRs) is slow primarily because of multistep downstream signaling. What is less well understood is the spatial and temporal kinetics of transmitter and receptor interaction. The present work uses the combination of the dopamine sensor, dLight, to detect the spatial release and diffusion of dopamine and a caged form of a D2-dopamine receptor antagonist, CyHQ-sulpiride, to rapidly block the D2 autoreceptors. Photoactivation of the CyHQ-sulpiride blocks receptors in milliseconds such that the time course of dopamine/receptor interaction is mapped onto the downstream signaling. The results show that highly localized release, but not dopamine diffusion, defines the time course of the functional interaction between dopamine and D2 autoreceptors, which determines downstream inhibition.
Collapse
Affiliation(s)
- Alec F Condon
- The Vollum Institute, Oregon Health Sciences University, Portland, OR, USA
| | - Brooks G Robinson
- The Vollum Institute, Oregon Health Sciences University, Portland, OR, USA
| | - Naeem Asad
- New York University Abu Dhabi, Saadiyat Island, P.O. Box 129188, Abu Dhabi, United Arab Emirates
| | - Timothy M Dore
- New York University Abu Dhabi, Saadiyat Island, P.O. Box 129188, Abu Dhabi, United Arab Emirates
| | - Lin Tian
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Davis, CA, USA
| | - John T Williams
- The Vollum Institute, Oregon Health Sciences University, Portland, OR, USA.
| |
Collapse
|
260
|
Limited functional convergence of eye-specific inputs in the retinogeniculate pathway of the mouse. Neuron 2021; 109:2457-2468.e12. [DOI: 10.1016/j.neuron.2021.05.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 04/16/2021] [Accepted: 05/28/2021] [Indexed: 11/22/2022]
|
261
|
δ-Protocadherins regulate neural progenitor cell division by antagonizing Ryk and Wnt/β-catenin signaling. iScience 2021; 24:102932. [PMID: 34430817 PMCID: PMC8374482 DOI: 10.1016/j.isci.2021.102932] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 05/10/2021] [Accepted: 07/28/2021] [Indexed: 12/24/2022] Open
Abstract
The division of neural progenitor cells provides the cellular substrate from which the nervous system is sculpted during development. The δ-protocadherin family of homophilic cell adhesion molecules is essential for the development of the vertebrate nervous system and is implicated in an array of neurodevelopmental disorders. We show that lesions in any of six, individual δ-protocadherins increases cell divisions of neural progenitors in the hindbrain. This increase is due to mis-regulation of Wnt/β-catenin signaling, as this pathway is upregulated in δ-protocadherin mutants and inhibition of this pathway blocks the increase in cell division. Furthermore, the δ-protocadherins can be present in complex with the Wnt receptor Ryk, and Ryk is required for the increased proliferation in protocadherin mutants. Thus, δ-protocadherins are novel regulators of Wnt/β-catenin signaling that may control the development of neural circuits by defining a molecular code for the identity of neural progenitor cells and differentially regulating their proliferation.
Collapse
|
262
|
Label-free two-photon imaging of mitochondrial activity in murine macrophages stimulated with bacterial and viral ligands. Sci Rep 2021; 11:14081. [PMID: 34234166 PMCID: PMC8263786 DOI: 10.1038/s41598-021-93043-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 06/16/2021] [Indexed: 11/12/2022] Open
Abstract
Mitochondria are the metabolic hub of the cell, playing a central role in regulating immune responses. Dysfunction of mitochondrial reprogramming can occur during bacterial and viral infections compromising hosts’ immune signaling. Comparative evaluation of these alterations in response to bacterial and viral ligands can provide insights into a cell’s ability to mount pathogen-specific responses. In this study, we used two-photon excitation fluorescence (TPEF) imaging to quantify reduced nicotinamide adenine dinucleotide phosphate (NAD(P)H) and flavin adenine dinucleotide (FAD) levels in the cell and to calculate the optical redox ratio (ORR), an indicator of mitochondrial dysfunction. Analyses were performed on RAW264.7 cells and murine bone marrow derived macrophages (BMM) stimulated with bacterial (LPS) and viral (Poly(I:C)) ligands. Responses were cell type dependent, with primary cells having significantly higher levels of FAD and higher oxygen consumption rates suggesting BMM may be more dependent on mitochondrial metabolism. Our findings also suggest that FAD-TPEF intensity may be a better predictor of mitochondrial activity and localization since it demonstrates unique mitochondrial clustering patterns in LPS vs. Poly(I:C) stimulated macrophages. Collectively, we demonstrate that TPEF imaging is a powerful label-free approach for quantifying changes in mitochondrial function and organization in macrophages following bacterial and viral stimuli.
Collapse
|
263
|
Glück C, Ferrari KD, Binini N, Keller A, Saab AS, Stobart JL, Weber B. Distinct signatures of calcium activity in brain mural cells. eLife 2021; 10:e70591. [PMID: 34227466 PMCID: PMC8294852 DOI: 10.7554/elife.70591] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 06/15/2021] [Indexed: 12/15/2022] Open
Abstract
Pericytes have been implicated in various neuropathologies, yet little is known about their function and signaling pathways in health. Here, we characterized calcium dynamics of cortical mural cells in anesthetized or awake Pdgfrb-CreERT2;Rosa26< LSL-GCaMP6s > mice and in acute brain slices. Smooth muscle cells (SMCs) and ensheathing pericytes (EPs), also named as terminal vascular SMCs, revealed similar calcium dynamics in vivo. In contrast, calcium signals in capillary pericytes (CPs) were irregular, higher in frequency, and occurred in cellular microdomains. In the absence of the vessel constricting agent U46619 in acute slices, SMCs and EPs revealed only sparse calcium signals, whereas CPs retained their spontaneous calcium activity. Interestingly, chemogenetic activation of neurons in vivo and acute elevations of extracellular potassium in brain slices strongly decreased calcium activity in CPs. We propose that neuronal activation and an extracellular increase in potassium suppress calcium activity in CPs, likely mediated by Kir2.2 and KATP channels.
Collapse
Affiliation(s)
- Chaim Glück
- Institute of Pharmacology and Toxicology, University of ZurichZürichSwitzerland
- Neuroscience Center Zurich, University and ETH ZurichZurichSwitzerland
| | - Kim David Ferrari
- Institute of Pharmacology and Toxicology, University of ZurichZürichSwitzerland
- Neuroscience Center Zurich, University and ETH ZurichZurichSwitzerland
| | - Noemi Binini
- Institute of Pharmacology and Toxicology, University of ZurichZürichSwitzerland
- Neuroscience Center Zurich, University and ETH ZurichZurichSwitzerland
| | - Annika Keller
- Neuroscience Center Zurich, University and ETH ZurichZurichSwitzerland
- Department of Neurosurgery, University of ZurichSchlierenSwitzerland
| | - Aiman S Saab
- Institute of Pharmacology and Toxicology, University of ZurichZürichSwitzerland
- Neuroscience Center Zurich, University and ETH ZurichZurichSwitzerland
| | - Jillian L Stobart
- Institute of Pharmacology and Toxicology, University of ZurichZürichSwitzerland
- Rady Faculty of Health Sciences, College of PharmacyWinnipegCanada
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of ZurichZürichSwitzerland
- Neuroscience Center Zurich, University and ETH ZurichZurichSwitzerland
| |
Collapse
|
264
|
Leighton AH, Cheyne JE, Houwen GJ, Maldonado PP, De Winter F, Levelt CN, Lohmann C. Somatostatin interneurons restrict cell recruitment to retinally driven spontaneous activity in the developing cortex. Cell Rep 2021; 36:109316. [PMID: 34233176 DOI: 10.1016/j.celrep.2021.109316] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 04/11/2021] [Accepted: 06/06/2021] [Indexed: 12/16/2022] Open
Abstract
During early development, before the eyes open, synaptic refinement of sensory networks depends on activity generated by developing neurons themselves. In the mouse visual system, retinal cells spontaneously depolarize and recruit downstream neurons to bursts of activity, where the number of recruited cells determines the resolution of synaptic retinotopic refinement. Here we show that during the second post-natal week in mouse visual cortex, somatostatin (SST)-expressing interneurons control the recruitment of cells to retinally driven spontaneous activity. Suppressing SST interneurons increases cell participation and allows events to spread farther along the cortex. During the same developmental period, a second type of high-participation, retina-independent event occurs. During these events, cells receive such large excitatory charge that inhibition is overwhelmed and large parts of the cortex participate in each burst. These results reveal a role of SST interneurons in restricting retinally driven activity in the visual cortex, which may contribute to the refinement of retinotopy.
Collapse
Affiliation(s)
- Alexandra H Leighton
- Department of Synapse and Network Development, Netherlands Institute for Neuroscience, 1105 BA Amsterdam, the Netherlands
| | - Juliette E Cheyne
- Department of Synapse and Network Development, Netherlands Institute for Neuroscience, 1105 BA Amsterdam, the Netherlands
| | - Gerrit J Houwen
- Department of Synapse and Network Development, Netherlands Institute for Neuroscience, 1105 BA Amsterdam, the Netherlands
| | - Paloma P Maldonado
- Department of Synapse and Network Development, Netherlands Institute for Neuroscience, 1105 BA Amsterdam, the Netherlands
| | - Fred De Winter
- Department of Neuroregeneration, Netherlands Institute for Neuroscience, 1105 BA Amsterdam, the Netherlands
| | - Christiaan N Levelt
- Department of Molecular Visual Plasticity, Netherlands Institute for Neuroscience, 1105 BA Amsterdam, the Netherlands; Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, Amsterdam, the Netherlands
| | - Christian Lohmann
- Department of Synapse and Network Development, Netherlands Institute for Neuroscience, 1105 BA Amsterdam, the Netherlands; Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
265
|
Shao LX, Liao C, Gregg I, Davoudian PA, Savalia NK, Delagarza K, Kwan AC. Psilocybin induces rapid and persistent growth of dendritic spines in frontal cortex in vivo. Neuron 2021; 109:2535-2544.e4. [PMID: 34228959 DOI: 10.1016/j.neuron.2021.06.008] [Citation(s) in RCA: 289] [Impact Index Per Article: 72.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 05/16/2021] [Accepted: 06/07/2021] [Indexed: 12/18/2022]
Abstract
Psilocybin is a serotonergic psychedelic with untapped therapeutic potential. There are hints that the use of psychedelics can produce neural adaptations, although the extent and timescale of the impact in a mammalian brain are unknown. In this study, we used chronic two-photon microscopy to image longitudinally the apical dendritic spines of layer 5 pyramidal neurons in the mouse medial frontal cortex. We found that a single dose of psilocybin led to ∼10% increases in spine size and density, driven by an elevated spine formation rate. The structural remodeling occurred quickly within 24 h and was persistent 1 month later. Psilocybin also ameliorated stress-related behavioral deficit and elevated excitatory neurotransmission. Overall, the results demonstrate that psilocybin-evoked synaptic rewiring in the cortex is fast and enduring, potentially providing a structural trace for long-term integration of experiences and lasting beneficial actions.
Collapse
Affiliation(s)
- Ling-Xiao Shao
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Clara Liao
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Ian Gregg
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Pasha A Davoudian
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT 06511, USA; Medical Scientist Training Program, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Neil K Savalia
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT 06511, USA; Medical Scientist Training Program, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Kristina Delagarza
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Alex C Kwan
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, USA; Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT 06511, USA; Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06511, USA.
| |
Collapse
|
266
|
Vishniakou I, Seelig JD. Differentiable model-based adaptive optics for two-photon microscopy. OPTICS EXPRESS 2021; 29:21418-21427. [PMID: 34265930 DOI: 10.1364/oe.424344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 06/01/2021] [Indexed: 06/13/2023]
Abstract
Aberrations limit scanning fluorescence microscopy when imaging in scattering materials such as biological tissue. Model-based approaches for adaptive optics take advantage of a computational model of the optical setup. Such models can be combined with the optimization techniques of machine learning frameworks to find aberration corrections, as was demonstrated for focusing a laser beam through aberrations onto a camera [Opt. Express2826436 (26436)10.1364/OE.403487]. Here, we extend this approach to two-photon scanning microscopy. The developed sensorless technique finds corrections for aberrations in scattering samples and will be useful for a range of imaging application, for example in brain tissue.
Collapse
|
267
|
Giardina G, Micko A, Bovenkamp D, Krause A, Placzek F, Papp L, Krajnc D, Spielvogel CP, Winklehner M, Höftberger R, Vila G, Andreana M, Leitgeb R, Drexler W, Wolfsberger S, Unterhuber A. Morpho-Molecular Metabolic Analysis and Classification of Human Pituitary Gland and Adenoma Biopsies Based on Multimodal Optical Imaging. Cancers (Basel) 2021; 13:3234. [PMID: 34209497 PMCID: PMC8267638 DOI: 10.3390/cancers13133234] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Pituitary adenomas count among the most common intracranial tumors. During pituitary oncogenesis structural, textural, metabolic and molecular changes occur which can be revealed with our integrated ultrahigh-resolution multimodal imaging approach including optical coherence tomography (OCT), multiphoton microscopy (MPM) and line scan Raman microspectroscopy (LSRM) on an unprecedented cellular level in a label-free manner. We investigated 5 pituitary gland and 25 adenoma biopsies, including lactotroph, null cell, gonadotroph, somatotroph and mammosomatotroph as well as corticotroph. First-level binary classification for discrimination of pituitary gland and adenomas was performed by feature extraction via radiomic analysis on OCT and MPM images and achieved an accuracy of 88%. Second-level multi-class classification was performed based on molecular analysis of the specimen via LSRM to discriminate pituitary adenomas subtypes with accuracies of up to 99%. Chemical compounds such as lipids, proteins, collagen, DNA and carotenoids and their relation could be identified as relevant biomarkers, and their spatial distribution visualized to provide deeper insight into the chemical properties of pituitary adenomas. Thereby, the aim of the current work was to assess a unique label-free and non-invasive multimodal optical imaging platform for pituitary tissue imaging and to perform a multiparametric morpho-molecular metabolic analysis and classification.
Collapse
Affiliation(s)
- Gabriel Giardina
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; (G.G.); (D.B.); (A.K.); (F.P.); (R.L.); (W.D.); (A.U.)
| | - Alexander Micko
- Department of Neurosurgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; (A.M.); (S.W.)
| | - Daniela Bovenkamp
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; (G.G.); (D.B.); (A.K.); (F.P.); (R.L.); (W.D.); (A.U.)
| | - Arno Krause
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; (G.G.); (D.B.); (A.K.); (F.P.); (R.L.); (W.D.); (A.U.)
| | - Fabian Placzek
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; (G.G.); (D.B.); (A.K.); (F.P.); (R.L.); (W.D.); (A.U.)
| | - Laszlo Papp
- QIMP Team, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; (L.P.); (D.K.)
| | - Denis Krajnc
- QIMP Team, Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; (L.P.); (D.K.)
| | - Clemens P. Spielvogel
- Christian Doppler Laboratory for Applied Metabolomics, Division of Nuclear Medicine, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria;
| | - Michael Winklehner
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; (M.W.); (R.H.)
| | - Romana Höftberger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; (M.W.); (R.H.)
| | - Greisa Vila
- Department of Internal Medicine III, Division of Endocrinology and Metabolism, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria;
| | - Marco Andreana
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; (G.G.); (D.B.); (A.K.); (F.P.); (R.L.); (W.D.); (A.U.)
| | - Rainer Leitgeb
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; (G.G.); (D.B.); (A.K.); (F.P.); (R.L.); (W.D.); (A.U.)
| | - Wolfgang Drexler
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; (G.G.); (D.B.); (A.K.); (F.P.); (R.L.); (W.D.); (A.U.)
| | - Stefan Wolfsberger
- Department of Neurosurgery, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; (A.M.); (S.W.)
| | - Angelika Unterhuber
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria; (G.G.); (D.B.); (A.K.); (F.P.); (R.L.); (W.D.); (A.U.)
| |
Collapse
|
268
|
Lohse M, Dahmen JC, Bajo VM, King AJ. Subcortical circuits mediate communication between primary sensory cortical areas in mice. Nat Commun 2021; 12:3916. [PMID: 34168153 PMCID: PMC8225818 DOI: 10.1038/s41467-021-24200-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 06/02/2021] [Indexed: 12/20/2022] Open
Abstract
Integration of information across the senses is critical for perception and is a common property of neurons in the cerebral cortex, where it is thought to arise primarily from corticocortical connections. Much less is known about the role of subcortical circuits in shaping the multisensory properties of cortical neurons. We show that stimulation of the whiskers causes widespread suppression of sound-evoked activity in mouse primary auditory cortex (A1). This suppression depends on the primary somatosensory cortex (S1), and is implemented through a descending circuit that links S1, via the auditory midbrain, with thalamic neurons that project to A1. Furthermore, a direct pathway from S1 has a facilitatory effect on auditory responses in higher-order thalamic nuclei that project to other brain areas. Crossmodal corticofugal projections to the auditory midbrain and thalamus therefore play a pivotal role in integrating multisensory signals and in enabling communication between different sensory cortical areas.
Collapse
Affiliation(s)
- Michael Lohse
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, UK.
- Sainsbury Wellcome Centre, London, UK.
| | - Johannes C Dahmen
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, UK
| | - Victoria M Bajo
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, UK
| | - Andrew J King
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
269
|
Jacob PF, Vargas-Gutierrez P, Okray Z, Vietti-Michelina S, Felsenberg J, Waddell S. Prior experience conditionally inhibits the expression of new learning in Drosophila. Curr Biol 2021; 31:3490-3503.e3. [PMID: 34146482 PMCID: PMC8409488 DOI: 10.1016/j.cub.2021.05.056] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/29/2021] [Accepted: 05/26/2021] [Indexed: 11/19/2022]
Abstract
Prior experience of a stimulus can inhibit subsequent acquisition or expression of a learned association of that stimulus. However, the neuronal manifestations of this learning effect, named latent inhibition (LI), are poorly understood. Here, we show that prior odor exposure can produce context-dependent LI of later appetitive olfactory memory performance in Drosophila. Odor pre-exposure forms a short-lived aversive memory whose lone expression lacks context-dependence. Acquisition of odor pre-exposure memory requires aversively reinforcing dopaminergic neurons that innervate two mushroom body compartments—one group of which exhibits increasing activity with successive odor experience. Odor-specific responses of the corresponding mushroom body output neurons are suppressed, and their output is necessary for expression of both pre-exposure memory and LI of appetitive memory. Therefore, odor pre-exposure attaches negative valence to the odor itself, and LI of appetitive memory results from a temporary and context-dependent retrieval deficit imposed by competition with the parallel short-lived aversive memory. Odor pre-exposure alters the expression of a learned association of that odor Pre-exposure memory only affects subsequent retrieval if context is consistent Pre-exposure memory can complement or compete with a learned association Odor pre-exposure forms a labile mushroom body-dependent aversive memory
Collapse
Affiliation(s)
- Pedro F Jacob
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford OX1 3TA, UK
| | | | - Zeynep Okray
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford OX1 3TA, UK
| | | | - Johannes Felsenberg
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford OX1 3TA, UK
| | - Scott Waddell
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford OX1 3TA, UK.
| |
Collapse
|
270
|
Rare CASP6N73T variant associated with hippocampal volume exhibits decreased proteolytic activity, synaptic transmission defect, and neurodegeneration. Sci Rep 2021; 11:12695. [PMID: 34135352 PMCID: PMC8209045 DOI: 10.1038/s41598-021-91367-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 05/25/2021] [Indexed: 01/22/2023] Open
Abstract
Caspase-6 (Casp6) is implicated in Alzheimer disease (AD) cognitive impairment and pathology. Hippocampal atrophy is associated with cognitive impairment in AD. Here, a rare functional exonic missense CASP6 single nucleotide polymorphism (SNP), causing the substitution of asparagine with threonine at amino acid 73 in Casp6 (Casp6N73T), was associated with hippocampal subfield CA1 volume preservation. Compared to wild type Casp6 (Casp6WT), recombinant Casp6N73T altered Casp6 proteolysis of natural substrates Lamin A/C and α-Tubulin, but did not alter cleavage of the Ac-VEID-AFC Casp6 peptide substrate. Casp6N73T-transfected HEK293T cells showed elevated Casp6 mRNA levels similar to Casp6WT-transfected cells, but, in contrast to Casp6WT, did not accumulate active Casp6 subunits nor show increased Casp6 enzymatic activity. Electrophysiological and morphological assessments showed that Casp6N73T recombinant protein caused less neurofunctional damage and neurodegeneration in hippocampal CA1 pyramidal neurons than Casp6WT. Lastly, CASP6 mRNA levels were increased in several AD brain regions confirming the implication of Casp6 in AD. These studies suggest that the rare Casp6N73T variant may protect against hippocampal atrophy due to its altered catalysis of natural protein substrates and intracellular instability thus leading to less Casp6-mediated damage to neuronal structure and function.
Collapse
|
271
|
Davidson AM, Mejía-Gómez H, Jacobowitz M, Mostany R. Dendritic Spine Density and Dynamics of Layer 5 Pyramidal Neurons of the Primary Motor Cortex Are Elevated With Aging. Cereb Cortex 2021; 30:767-777. [PMID: 31298696 DOI: 10.1093/cercor/bhz124] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 04/17/2019] [Accepted: 05/20/2019] [Indexed: 01/04/2023] Open
Abstract
It is well established that motor impairment often occurs alongside healthy aging, leading to problems with fine motor skills and coordination. Although previously thought to be caused by neuronal death accumulating across the lifespan, it is now believed that the source of this impairment instead stems from more subtle changes in neural connectivity. The dendritic spine is a prime target for exploration of this problem because it is the postsynaptic partner of most excitatory synapses received by the pyramidal neuron, a cortical cell that carries much of the information processing load in the cerebral cortex. We repeatedly imaged the same dendrites in young adult and aged mouse motor cortex over the course of 1 month to look for differences in the baseline state of the dendritic spine population. These experiments reveal increased dendritic spine density, without obvious changes in spine clustering, occurring at the aged dendrite. Additionally, aged dendrites exhibit elevated spine turnover and stabilization alongside decreased long-term spine survival. These results suggest that at baseline the aged motor cortex may exist in a perpetual state of relative instability and attempts at compensation. This phenotype of aging may provide clues for future targets of aging-related motor impairment remediation.
Collapse
Affiliation(s)
- A M Davidson
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA.,Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - H Mejía-Gómez
- Neuroscience Program, Brain Institute, Tulane University, New Orleans, LA 70118, USA.,Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - M Jacobowitz
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - R Mostany
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA.,Brain Institute, Tulane University, New Orleans, LA 70118, USA
| |
Collapse
|
272
|
Pilger C, Pospíšil J, Müller M, Ruoff M, Schütte M, Spiecker H, Huser T. Super-resolution fluorescence microscopy by line-scanning with an unmodified two-photon microscope. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2021; 379:20200300. [PMID: 33896201 PMCID: PMC8072199 DOI: 10.1098/rsta.2020.0300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/20/2020] [Indexed: 05/19/2023]
Abstract
Fluorescence-based microscopy as one of the standard tools in biomedical research benefits more and more from super-resolution methods, which offer enhanced spatial resolution allowing insights into new biological processes. A typical drawback of using these methods is the need for new, complex optical set-ups. This becomes even more significant when using two-photon fluorescence excitation, which offers deep tissue imaging and excellent z-sectioning. We show that the generation of striped-illumination patterns in two-photon laser scanning microscopy can readily be exploited for achieving optical super-resolution and contrast enhancement using open-source image reconstruction software. The special appeal of this approach is that even in the case of a commercial two-photon laser scanning microscope no optomechanical modifications are required to achieve this modality. Modifying the scanning software with a custom-written macro to address the scanning mirrors in combination with rapid intensity switching by an electro-optic modulator is sufficient to accomplish the acquisition of two-photon striped-illumination patterns on an sCMOS camera. We demonstrate and analyse the resulting resolution improvement by applying different recently published image resolution evaluation procedures to the reconstructed filtered widefield and super-resolved images. This article is part of the Theo Murphy meeting issue 'Super-resolution structured illumination microscopy (part 1)'.
Collapse
Affiliation(s)
- Christian Pilger
- Biomolecular Photonics, Department of Physics, University of Bielefeld, Bielefeld, Germany
| | - Jakub Pospíšil
- Biomolecular Photonics, Department of Physics, University of Bielefeld, Bielefeld, Germany
- Department of Radioelectronics, Faculty of Electrical Engineering, Czech Technical University in Prague, Technická 2, 166 27 Prague 6, Czech Republic
| | - Marcel Müller
- Biomolecular Photonics, Department of Physics, University of Bielefeld, Bielefeld, Germany
| | - Martin Ruoff
- LaVision BioTec GmbH, Astastraße 14, 33617 Bielefeld, Germany
| | - Martin Schütte
- LaVision BioTec GmbH, Astastraße 14, 33617 Bielefeld, Germany
| | | | - Thomas Huser
- Biomolecular Photonics, Department of Physics, University of Bielefeld, Bielefeld, Germany
| |
Collapse
|
273
|
Orsolic I, Rio M, Mrsic-Flogel TD, Znamenskiy P. Mesoscale cortical dynamics reflect the interaction of sensory evidence and temporal expectation during perceptual decision-making. Neuron 2021; 109:1861-1875.e10. [PMID: 33861941 PMCID: PMC8186564 DOI: 10.1016/j.neuron.2021.03.031] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 01/17/2021] [Accepted: 03/25/2021] [Indexed: 01/01/2023]
Abstract
How sensory evidence is transformed across multiple brain regions to influence behavior remains poorly understood. We trained mice in a visual change detection task designed to separate the covert antecedents of choices from activity associated with their execution. Wide-field calcium imaging across the dorsal cortex revealed fundamentally different dynamics of activity underlying these processes. Although signals related to execution of choice were widespread, fluctuations in sensory evidence in the absence of overt motor responses triggered a confined activity cascade, beginning with transient modulation of visual cortex and followed by sustained recruitment of the secondary and primary motor cortex. Activation of the motor cortex by sensory evidence was modulated by animals' expectation of when the stimulus was likely to change. These results reveal distinct activation timescales of specific cortical areas by sensory evidence during decision-making and show that recruitment of the motor cortex depends on the interaction of sensory evidence and temporal expectation.
Collapse
Affiliation(s)
- Ivana Orsolic
- Sainsbury Wellcome Centre, University College London, 25 Howland Street, London W1T 4JG, UK; Biozentrum, University of Basel, Klingelbergstrasse 70, 4056 Basel, Switzerland
| | - Maxime Rio
- Sainsbury Wellcome Centre, University College London, 25 Howland Street, London W1T 4JG, UK; Biozentrum, University of Basel, Klingelbergstrasse 70, 4056 Basel, Switzerland; The National Institute of Water and Atmospheric Research, 301 Evans Bay Parade, Hataitai, Wellington 6021, New Zealand
| | - Thomas D Mrsic-Flogel
- Sainsbury Wellcome Centre, University College London, 25 Howland Street, London W1T 4JG, UK; Biozentrum, University of Basel, Klingelbergstrasse 70, 4056 Basel, Switzerland.
| | - Petr Znamenskiy
- Sainsbury Wellcome Centre, University College London, 25 Howland Street, London W1T 4JG, UK; Biozentrum, University of Basel, Klingelbergstrasse 70, 4056 Basel, Switzerland; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
274
|
Ketamine Rapidly Enhances Glutamate-Evoked Dendritic Spinogenesis in Medial Prefrontal Cortex Through Dopaminergic Mechanisms. Biol Psychiatry 2021; 89:1096-1105. [PMID: 33637303 PMCID: PMC8740507 DOI: 10.1016/j.biopsych.2020.12.022] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/14/2020] [Accepted: 12/28/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Ketamine elicits rapid onset antidepressant effects in patients with clinical depression through mechanisms hypothesized to involve the genesis of neocortical dendritic spines and synapses. Yet, the observed changes in dendritic spine morphology usually emerge well after ketamine clearance, raising questions about the link between rapid behavioral effects of ketamine and plasticity. METHODS Here, we used two-photon glutamate uncaging/imaging to focally induce spinogenesis in the medial prefrontal cortex, directly interrogating baseline and ketamine-associated plasticity of deep layer pyramidal neurons in C57BL/6 mice. We combined pharmacological, genetic, optogenetic, and chemogenetic manipulations to interrogate dopaminergic mechanisms underlying ketamine-induced rapid enhancement in evoked plasticity and associated behavioral changes. RESULTS We found that ketamine rapidly enhances glutamate-evoked spinogenesis in the medial prefrontal cortex, with timing that matches the onset of its behavioral efficacy and precedes changes in dendritic spine density. Ketamine increases evoked cortical spinogenesis through dopamine Drd1 receptor (Drd1) activation that requires dopamine release, compensating blunted plasticity in a learned helplessness paradigm. The enhancement in evoked spinogenesis after Drd1 activation or ketamine treatment depends on postsynaptic protein kinase A activity. Furthermore, ketamine's behavioral effects are blocked by chemogenetic inhibition of dopamine release and mimicked by activating presynaptic dopaminergic terminals or postsynaptic Gαs-coupled cascades in the medial prefrontal cortex. CONCLUSIONS Our findings highlight dopaminergic mediation of rapid enhancement in activity-dependent dendritic spinogenesis and behavioral effects induced by ketamine.
Collapse
|
275
|
Inhibitory regulation of calcium transients in prefrontal dendritic spines is compromised by a nonsense Shank3 mutation. Mol Psychiatry 2021; 26:1945-1966. [PMID: 32161363 PMCID: PMC7483244 DOI: 10.1038/s41380-020-0708-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 02/23/2020] [Accepted: 02/28/2020] [Indexed: 01/11/2023]
Abstract
The SHANK3 gene encodes a postsynaptic scaffold protein in excitatory synapses, and its disruption is implicated in neurodevelopmental disorders such as Phelan-McDermid syndrome, autism spectrum disorder, and schizophrenia. Most studies of SHANK3 in the neocortex and hippocampus have focused on disturbances in pyramidal neurons. However, GABAergic interneurons likewise receive excitatory inputs and presumably would also be a target of constitutive SHANK3 perturbations. In this study, we characterize the prefrontal cortical microcircuit in awake mice using subcellular-resolution two-photon microscopy. We focused on a nonsense R1117X mutation, which leads to truncated SHANK3 and has been linked previously to cortical dysfunction. We find that R1117X mutants have abnormally elevated calcium transients in apical dendritic spines. The synaptic calcium dysregulation is due to a loss of dendritic inhibition via decreased NMDAR currents and reduced firing of dendrite-targeting somatostatin-expressing (SST) GABAergic interneurons. Notably, upregulation of the NMDAR subunit GluN2B in SST interneurons corrects the excessive synaptic calcium signals and ameliorates learning deficits in R1117X mutants. These findings reveal dendrite-targeting interneurons, and more broadly the inhibitory control of dendritic spines, as a key microcircuit mechanism compromised by the SHANK3 dysfunction.
Collapse
|
276
|
Maltese M, March JR, Bashaw AG, Tritsch NX. Dopamine differentially modulates the size of projection neuron ensembles in the intact and dopamine-depleted striatum. eLife 2021; 10:e68041. [PMID: 33983121 PMCID: PMC8163504 DOI: 10.7554/elife.68041] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/12/2021] [Indexed: 12/20/2022] Open
Abstract
Dopamine (DA) is a critical modulator of brain circuits that control voluntary movements, but our understanding of its influence on the activity of target neurons in vivo remains limited. Here, we use two-photon Ca2+ imaging to monitor the activity of direct and indirect-pathway spiny projection neurons (SPNs) simultaneously in the striatum of behaving mice during acute and prolonged manipulations of DA signaling. We find that increasing and decreasing DA biases striatal activity toward the direct and indirect pathways, respectively, by changing the overall number of SPNs recruited during behavior in a manner not predicted by existing models of DA function. This modulation is drastically altered in a model of Parkinson's disease. Our results reveal a previously unappreciated population-level influence of DA on striatal output and provide novel insights into the pathophysiology of Parkinson's disease.
Collapse
Affiliation(s)
- Marta Maltese
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
- Fresco Institute for Parkinson’s and Movement Disorders, New York University Langone HealthNew YorkUnited States
| | - Jeffrey R March
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
- Fresco Institute for Parkinson’s and Movement Disorders, New York University Langone HealthNew YorkUnited States
| | - Alexander G Bashaw
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
- Fresco Institute for Parkinson’s and Movement Disorders, New York University Langone HealthNew YorkUnited States
| | - Nicolas X Tritsch
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
- Fresco Institute for Parkinson’s and Movement Disorders, New York University Langone HealthNew YorkUnited States
| |
Collapse
|
277
|
Fulton KA, Briggman KL. Permeabilization-free en bloc immunohistochemistry for correlative microscopy. eLife 2021; 10:63392. [PMID: 33983117 PMCID: PMC8118656 DOI: 10.7554/elife.63392] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 04/29/2021] [Indexed: 01/03/2023] Open
Abstract
A dense reconstruction of neuronal synaptic connectivity typically requires high-resolution 3D electron microscopy (EM) data, but EM data alone lacks functional information about neurons and synapses. One approach to augment structural EM datasets is with the fluorescent immunohistochemical (IHC) localization of functionally relevant proteins. We describe a protocol that obviates the requirement of tissue permeabilization in thick tissue sections, a major impediment for correlative pre-embedding IHC and EM. We demonstrate the permeabilization-free labeling of neuronal cell types, intracellular enzymes, and synaptic proteins in tissue sections hundreds of microns thick in multiple brain regions from mice while simultaneously retaining the ultrastructural integrity of the tissue. Finally, we explore the utility of this protocol by performing proof-of-principle correlative experiments combining two-photon imaging of protein distributions and 3D EM.
Collapse
Affiliation(s)
- Kara A Fulton
- Brown University, Providence, United States.,National Institute of Neurological Disorders and Stroke (NINDS), Bethesda, United States.,Center of Advanced European Studies and Research (caesar), Bonn, Germany
| | - Kevin L Briggman
- National Institute of Neurological Disorders and Stroke (NINDS), Bethesda, United States.,Center of Advanced European Studies and Research (caesar), Bonn, Germany
| |
Collapse
|
278
|
Efficient optogenetic silencing of neurotransmitter release with a mosquito rhodopsin. Neuron 2021; 109:1621-1635.e8. [PMID: 33979634 DOI: 10.1016/j.neuron.2021.03.013] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/02/2021] [Accepted: 03/09/2021] [Indexed: 12/21/2022]
Abstract
Information is carried between brain regions through neurotransmitter release from axonal presynaptic terminals. Understanding the functional roles of defined neuronal projection pathways requires temporally precise manipulation of their activity. However, existing inhibitory optogenetic tools have low efficacy and off-target effects when applied to presynaptic terminals, while chemogenetic tools are difficult to control in space and time. Here, we show that a targeting-enhanced mosquito homolog of the vertebrate encephalopsin (eOPN3) can effectively suppress synaptic transmission through the Gi/o signaling pathway. Brief illumination of presynaptic terminals expressing eOPN3 triggers a lasting suppression of synaptic output that recovers spontaneously within minutes in vitro and in vivo. In freely moving mice, eOPN3-mediated suppression of dopaminergic nigrostriatal afferents induces a reversible ipsiversive rotational bias. We conclude that eOPN3 can be used to selectively suppress neurotransmitter release at presynaptic terminals with high spatiotemporal precision, opening new avenues for functional interrogation of long-range neuronal circuits in vivo.
Collapse
|
279
|
Blot A, Roth MM, Gasler I, Javadzadeh M, Imhof F, Hofer SB. Visual intracortical and transthalamic pathways carry distinct information to cortical areas. Neuron 2021; 109:1996-2008.e6. [PMID: 33979633 PMCID: PMC8221812 DOI: 10.1016/j.neuron.2021.04.017] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 02/28/2021] [Accepted: 04/15/2021] [Indexed: 01/13/2023]
Abstract
Sensory processing involves information flow between neocortical areas, assumed to rely on direct intracortical projections. However, cortical areas may also communicate indirectly via higher-order nuclei in the thalamus, such as the pulvinar or lateral posterior nucleus (LP) in the visual system of rodents. The fine-scale organization and function of these cortico-thalamo-cortical pathways remains unclear. We find that responses of mouse LP neurons projecting to higher visual areas likely derive from feedforward input from primary visual cortex (V1) combined with information from many cortical and subcortical areas, including superior colliculus. Signals from LP projections to different higher visual areas are tuned to specific features of visual stimuli and their locomotor context, distinct from the signals carried by direct intracortical projections from V1. Thus, visual transthalamic pathways are functionally specific to their cortical target, different from feedforward cortical pathways, and combine information from multiple brain regions, linking sensory signals with behavioral context. Transthalamic pathway through pulvinar indirectly connects lower to higher cortical areas This pathway combines input from V1 with that of many cortical and subcortical areas Pulvinar conveys distinct visual and motor information to different higher visual areas Direct intracortical and transthalamic pathways convey different information
Collapse
Affiliation(s)
- Antonin Blot
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, London, UK; Biozentrum, University of Basel, Basel, Switzerland
| | | | - Ioana Gasler
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, London, UK; Biozentrum, University of Basel, Basel, Switzerland
| | - Mitra Javadzadeh
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, London, UK; Biozentrum, University of Basel, Basel, Switzerland
| | - Fabia Imhof
- Biozentrum, University of Basel, Basel, Switzerland
| | - Sonja B Hofer
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College London, London, UK; Biozentrum, University of Basel, Basel, Switzerland.
| |
Collapse
|
280
|
Barz CS, Garderes PM, Ganea DA, Reischauer S, Feldmeyer D, Haiss F. Functional and Structural Properties of Highly Responsive Somatosensory Neurons in Mouse Barrel Cortex. Cereb Cortex 2021; 31:4533-4553. [PMID: 33963394 PMCID: PMC8408454 DOI: 10.1093/cercor/bhab104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 03/12/2021] [Accepted: 03/24/2021] [Indexed: 11/14/2022] Open
Abstract
Sparse population activity is a well-known feature of supragranular sensory neurons in neocortex. The mechanisms underlying sparseness are not well understood because a direct link between the neurons activated in vivo, and their cellular properties investigated in vitro has been missing. We used two-photon calcium imaging to identify a subset of neurons in layer L2/3 (L2/3) of mouse primary somatosensory cortex that are highly active following principal whisker vibrotactile stimulation. These high responders (HRs) were then tagged using photoconvertible green fluorescent protein for subsequent targeting in the brain slice using intracellular patch-clamp recordings and biocytin staining. This approach allowed us to investigate the structural and functional properties of HRs that distinguish them from less active control cells. Compared to less responsive L2/3 neurons, HRs displayed increased levels of stimulus-evoked and spontaneous activity, elevated noise and spontaneous pairwise correlations, and stronger coupling to the population response. Intrinsic excitability was reduced in HRs, while we found no evidence for differences in other electrophysiological and morphological parameters. Thus, the choice of which neurons participate in stimulus encoding may be determined largely by network connectivity rather than by cellular structure and function.
Collapse
Affiliation(s)
- C S Barz
- Institute of Neuroscience and Medicine, INM-10, Research Centre Jülich, 52425 Jülich, Germany.,Department of Psychiatry, Psychotherapy and Psychosomatics, Medical School, RWTH Aachen University, 52074 Aachen, Germany.,Jülich-Aachen Research Alliance - Translational Brain Medicine, 52074 Aachen, Germany.,IZKF Aachen, Medical School, RWTH Aachen University, 52074 Aachen, Germany
| | - P M Garderes
- IZKF Aachen, Medical School, RWTH Aachen University, 52074 Aachen, Germany.,Department of Neuropathology, Medical School, RWTH Aachen University, 52074 Aachen, Germany.,Department of Ophthalmology, Medical School, RWTH Aachen University, 52074 Aachen, Germany.,Unit of Neural Circuits Dynamics and Decision Making, Institut Pasteur, 75015 Paris, France
| | - D A Ganea
- IZKF Aachen, Medical School, RWTH Aachen University, 52074 Aachen, Germany.,Department of Neuropathology, Medical School, RWTH Aachen University, 52074 Aachen, Germany.,Department of Ophthalmology, Medical School, RWTH Aachen University, 52074 Aachen, Germany.,Biomedical Department, University of Basel, 4056 Basel, Switzerland
| | - S Reischauer
- Medical Clinic I, (Cardiology/Angiology) and Campus Kerckhoff, Justus-Liebig-University Giessen, 35390 Giessen Germany.,Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany.,Cardio-Pulmonary Institute (CPI), 35392 Giessen, Germany
| | - D Feldmeyer
- Institute of Neuroscience and Medicine, INM-10, Research Centre Jülich, 52425 Jülich, Germany.,Department of Psychiatry, Psychotherapy and Psychosomatics, Medical School, RWTH Aachen University, 52074 Aachen, Germany.,Jülich-Aachen Research Alliance - Translational Brain Medicine, 52074 Aachen, Germany
| | - F Haiss
- IZKF Aachen, Medical School, RWTH Aachen University, 52074 Aachen, Germany.,Department of Neuropathology, Medical School, RWTH Aachen University, 52074 Aachen, Germany.,Department of Ophthalmology, Medical School, RWTH Aachen University, 52074 Aachen, Germany.,Unit of Neural Circuits Dynamics and Decision Making, Institut Pasteur, 75015 Paris, France
| |
Collapse
|
281
|
Yao J, Sun B, Institoris A, Zhan X, Guo W, Song Z, Liu Y, Hiess F, Boyce AKJ, Ni M, Wang R, Ter Keurs H, Back TG, Fill M, Thompson RJ, Turner RW, Gordon GR, Chen SRW. Limiting RyR2 Open Time Prevents Alzheimer's Disease-Related Neuronal Hyperactivity and Memory Loss but Not β-Amyloid Accumulation. Cell Rep 2021; 32:108169. [PMID: 32966798 PMCID: PMC7532726 DOI: 10.1016/j.celrep.2020.108169] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 07/23/2020] [Accepted: 08/27/2020] [Indexed: 12/31/2022] Open
Abstract
Neuronal hyperactivity is an early primary dysfunction in Alzheimer’s disease (AD) in humans and animal models, but effective neuronal hyperactivity-directed anti-AD therapeutic agents are lacking. Here we define a previously unknown mode of ryanodine receptor 2 (RyR2) control of neuronal hyperactivity and AD progression. We show that a single RyR2 point mutation, E4872Q, which reduces RyR2 open time, prevents hyperexcitability, hyperactivity, memory impairment, neuronal cell death, and dendritic spine loss in a severe early-onset AD mouse model (5xFAD). The RyR2-E4872Q mutation upregulates hippocampal CA1-pyramidal cell A-type K+ current, a well-known neuronal excitability control that is downregulated in AD. Pharmacologically limiting RyR2 open time with the R-carvedilol enantiomer (but not racemic carvedilol) prevents and rescues neuronal hyperactivity, memory impairment, and neuron loss even in late stages of AD. These AD-related deficits are prevented even with continued β-amyloid accumulation. Thus, limiting RyR2 open time may be a hyperactivity-directed, non-β-amyloid-targeted anti-AD strategy. Yao et al. show that genetically or pharmacologically limiting the open duration of ryanodine receptor 2 upregulates the A-type potassium current and prevents neuronal hyperexcitability and hyperactivity, memory impairment, neuronal cell death, and dendritic spine loss in a severe early-onset Alzheimer’s disease mouse model, even with continued accumulation of β-amyloid.
Collapse
Affiliation(s)
- Jinjing Yao
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Bo Sun
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada; Medical School, Kunming University of Science and Technology, Kunming 650504, China
| | - Adam Institoris
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Xiaoqin Zhan
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Wenting Guo
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Zhenpeng Song
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Yajing Liu
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Florian Hiess
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Andrew K J Boyce
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Mingke Ni
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Ruiwu Wang
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Henk Ter Keurs
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Thomas G Back
- Department of Chemistry, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Michael Fill
- Department of Physiology & Biophysics, Rush University Medical Center, Chicago, IL 60612, USA
| | - Roger J Thompson
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Ray W Turner
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Grant R Gordon
- Hotchkiss Brain Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - S R Wayne Chen
- Libin Cardiovascular Institute, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada; Department of Physiology & Biophysics, Rush University Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
282
|
Liu Y, Xin Y, Xu NL. A cortical circuit mechanism for structural knowledge-based flexible sensorimotor decision-making. Neuron 2021; 109:2009-2024.e6. [PMID: 33957065 DOI: 10.1016/j.neuron.2021.04.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 03/01/2021] [Accepted: 04/14/2021] [Indexed: 10/21/2022]
Abstract
Making flexible decisions based on prior knowledge about causal environmental structures is a hallmark of goal-directed cognition in mammalian brains. Although several association brain regions, including the orbitofrontal cortex (OFC), have been implicated, the precise neuronal circuit mechanisms underlying knowledge-based decision-making remain elusive. Here, we established an inference-based auditory categorization task where mice performed within-session flexible stimulus re-categorization by inferring the changing task rules. We constructed a reinforcement learning model to recapitulate the inference-based flexible behavior and quantify the hidden variables associated with task structural knowledge. Combining two-photon population imaging and projection-specific optogenetics, we found that auditory cortex (ACx) neurons encoded the hidden task rule variable, which requires feedback input from the OFC. Silencing OFC-ACx input specifically disrupted re-categorization behavior. Direct imaging from OFC axons in the ACx revealed task state-related feedback signals, supporting the knowledge-based updating mechanism. Our data reveal a cortical circuit mechanism underlying structural knowledge-based flexible decision-making.
Collapse
Affiliation(s)
- Yanhe Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Yu Xin
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Ning-Long Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of the Chinese Academy of Sciences, Beijing 100049, China; Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai 201210, China.
| |
Collapse
|
283
|
Altered Phosphorylation of the Proteasome Subunit Rpt6 Has Minimal Impact on Synaptic Plasticity and Learning. eNeuro 2021; 8:ENEURO.0073-20.2021. [PMID: 33658307 PMCID: PMC8116113 DOI: 10.1523/eneuro.0073-20.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 01/13/2021] [Accepted: 01/27/2021] [Indexed: 11/21/2022] Open
Abstract
Dynamic control of protein degradation via the ubiquitin proteasome system (UPS) is thought to play a crucial role in neuronal function and synaptic plasticity. The proteasome subunit Rpt6, an AAA ATPase subunit of the 19S regulatory particle (RP), has emerged as an important site for regulation of 26S proteasome function in neurons. Phosphorylation of Rpt6 on serine 120 (S120) can stimulate the catalytic rate of substrate degradation by the 26S proteasome and this site is targeted by the plasticity-related kinase Ca2+/calmodulin-dependent kinase II (CaMKII), making it an attractive candidate for regulation of proteasome function in neurons. Several in vitro studies have shown that altered Rpt6 S120 phosphorylation can affect the structure and function of synapses. To evaluate the importance of Rpt6 S120 phosphorylation in vivo, we created two mouse models which feature mutations at S120 that block or mimic phosphorylation at this site. We find that peptidase and ATPase activities are upregulated in the phospho-mimetic mutant and downregulated in the phospho-dead mutant [S120 mutated to aspartic acid (S120D) or alanine (S120A), respectively]. Surprisingly, these mutations had no effect on basal synaptic transmission, long-term potentiation (LTP), and dendritic spine dynamics and density in the hippocampus. Furthermore, these mutants displayed no deficits in cued and contextual fear memory. Thus, in a mouse model that blocks or mimics phosphorylation at this site, either compensatory mechanisms negate these effects, or small variations in proteasome activity are not enough to induce significant changes in synaptic structure, plasticity, or behavior.
Collapse
|
284
|
El-Quessny M, Maanum K, Feller MB. Visual Experience Influences Dendritic Orientation but Is Not Required for Asymmetric Wiring of the Retinal Direction Selective Circuit. Cell Rep 2021; 31:107844. [PMID: 32610144 DOI: 10.1016/j.celrep.2020.107844] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 05/22/2020] [Accepted: 06/10/2020] [Indexed: 01/02/2023] Open
Abstract
Changes in dendritic morphology in response to activity have long been thought to be a critical component of how neural circuits develop to properly encode sensory information. Ventral-preferring direction-selective ganglion cells (vDSGCs) have asymmetric dendrites oriented along their preferred direction, and this has been hypothesized to play a critical role in their tuning. Here we report the surprising result that visual experience is critical for the alignment of vDSGC dendrites to their preferred direction. Interestingly, vDSGCs in dark-reared mice lose their inhibition-independent dendritic contribution to direction-selective tuning while maintaining asymmetric inhibitory input. These data indicate that different mechanisms of a cell's computational abilities can be constructed over development through divergent mechanisms.
Collapse
Affiliation(s)
- Malak El-Quessny
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kayla Maanum
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Marla B Feller
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
285
|
Wu M, Minkowicz S, Dumrongprechachan V, Hamilton P, Xiao L, Kozorovitskiy Y. Attenuated dopamine signaling after aversive learning is restored by ketamine to rescue escape actions. eLife 2021; 10:64041. [PMID: 33904412 PMCID: PMC8211450 DOI: 10.7554/elife.64041] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 04/26/2021] [Indexed: 12/13/2022] Open
Abstract
Escaping aversive stimuli is essential for complex organisms, but prolonged exposure to stress leads to maladaptive learning. Stress alters neuronal activity and neuromodulatory signaling in distributed networks, modifying behavior. Here, we describe changes in dopaminergic neuron activity and signaling following aversive learning in a learned helplessness paradigm in mice. A single dose of ketamine suffices to restore escape behavior after aversive learning. Dopaminergic neuron activity in the ventral tegmental area (VTA) systematically varies across learning, correlating with future sensitivity to ketamine treatment. Ketamine’s effects are blocked by chemogenetic inhibition of dopamine signaling. Rather than directly altering the activity of dopaminergic neurons, ketamine appears to rescue dopamine dynamics through actions in the medial prefrontal cortex (mPFC). Chemogenetic activation of Drd1 receptor positive mPFC neurons mimics ketamine’s effects on behavior. Together, our data link neuromodulatory dynamics in mPFC-VTA circuits, aversive learning, and the effects of ketamine. Over 264 million people around the world suffer from depression, according to the World Health Organization (WHO). Depression can be debilitating, and while anti-depressant drugs are available, they do not always work. A small molecule drug mainly used for anesthesia called ketamine has recently been shown to ameliorate depressive symptoms within hours, much faster than most anti-depressants. However, the molecular mechanisms behind this effect are still largely unknown. Most anti-depressant drugs work by restoring the normal balance of dopamine and other chemical messengers in the brain. Dopamine is released by a specialized group of cells called dopaminergic neurons, and helps us make decisions by influencing a wide range of other cells in the brain. In a healthy brain, dopamine directs us to rewarding choices, while avoiding actions with negative outcomes. During depression, these dopamine signals are perturbed, resulting in reduced motivation and pleasure. But it remained unclear whether ketamine’s anti-depressant activity also relied on dopamine. To investigate this, Wu et al. used a behavioral study called “learned helplessness” which simulates depression by putting mice in unavoidable stressful situations. Over time the mice learn that their actions do not change the outcome and eventually stop trying to escape from unpleasant situations, even if they are avoidable. The experiment showed that dopaminergic neurons in an area of the brain that is an important part of the “reward and aversion” system became less sensitive to unpleasant stimuli following learned helplessness. When the mice received ketamine, these neurons recovered after a few hours. Individual mice also responded differently to ketamine. The most ‘resilient’, stress-resistant mice, which had distinct patterns of dopamine signaling, also responded most strongly to the drug. Genetic and chemical manipulation of dopaminergic neurons confirmed that ketamine needed intact dopamine signals to work, and revealed that it acted indirectly on dopamine dynamics via another brain region called the medial prefrontal cortex. These results shed new light on how a promising new anti-depressant works. In the future, they may also explain why drugs like ketamine work better for some people than others, ultimately helping clinicians select the most effective treatment for individual patients.
Collapse
Affiliation(s)
- Mingzheng Wu
- Department of Neurobiology, Northwestern University, Evanston, United States
| | - Samuel Minkowicz
- Department of Neurobiology, Northwestern University, Evanston, United States
| | | | - Pauline Hamilton
- Department of Neurobiology, Northwestern University, Evanston, United States
| | - Lei Xiao
- Department of Neurobiology, Northwestern University, Evanston, United States
| | | |
Collapse
|
286
|
Reinert S, Hübener M, Bonhoeffer T, Goltstein PM. Mouse prefrontal cortex represents learned rules for categorization. Nature 2021; 593:411-417. [PMID: 33883745 PMCID: PMC8131197 DOI: 10.1038/s41586-021-03452-z] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 03/12/2021] [Indexed: 12/03/2022]
Abstract
The ability to categorize sensory stimuli is crucial for an animal’s survival in a complex environment. Memorizing categories instead of individual exemplars enables greater behavioural flexibility and is computationally advantageous. Neurons that show category selectivity have been found in several areas of the mammalian neocortex1–4, but the prefrontal cortex seems to have a prominent role4,5 in this context. Specifically, in primates that are extensively trained on a categorization task, neurons in the prefrontal cortex rapidly and flexibly represent learned categories6,7. However, how these representations first emerge in naive animals remains unexplored, leaving it unclear whether flexible representations are gradually built up as part of semantic memory or assigned more or less instantly during task execution8,9. Here we investigate the formation of a neuronal category representation throughout the entire learning process by repeatedly imaging individual cells in the mouse medial prefrontal cortex. We show that mice readily learn rule-based categorization and generalize to novel stimuli. Over the course of learning, neurons in the prefrontal cortex display distinct dynamics in acquiring category selectivity and are differentially engaged during a later switch in rules. A subset of neurons selectively and uniquely respond to categories and reflect generalization behaviour. Thus, a category representation in the mouse prefrontal cortex is gradually acquired during learning rather than recruited ad hoc. This gradual process suggests that neurons in the medial prefrontal cortex are part of a specific semantic memory for visual categories. Neurons in the mouse medial prefrontal cortex acquire category-selective responses with learning.
Collapse
Affiliation(s)
- Sandra Reinert
- Max Planck Institute of Neurobiology, Martinsried, Germany.,Graduate School of Systemic Neurosciences, Ludwig-Maximilians-Universität München, Martinsried, Germany
| | - Mark Hübener
- Max Planck Institute of Neurobiology, Martinsried, Germany
| | | | | |
Collapse
|
287
|
Stringer C, Michaelos M, Tsyboulski D, Lindo SE, Pachitariu M. High-precision coding in visual cortex. Cell 2021; 184:2767-2778.e15. [PMID: 33857423 DOI: 10.1016/j.cell.2021.03.042] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 03/04/2021] [Accepted: 03/19/2021] [Indexed: 01/18/2023]
Abstract
Individual neurons in visual cortex provide the brain with unreliable estimates of visual features. It is not known whether the single-neuron variability is correlated across large neural populations, thus impairing the global encoding of stimuli. We recorded simultaneously from up to 50,000 neurons in mouse primary visual cortex (V1) and in higher order visual areas and measured stimulus discrimination thresholds of 0.35° and 0.37°, respectively, in an orientation decoding task. These neural thresholds were almost 100 times smaller than the behavioral discrimination thresholds reported in mice. This discrepancy could not be explained by stimulus properties or arousal states. Furthermore, behavioral variability during a sensory discrimination task could not be explained by neural variability in V1. Instead, behavior-related neural activity arose dynamically across a network of non-sensory brain areas. These results imply that perceptual discrimination in mice is limited by downstream decoders, not by neural noise in sensory representations.
Collapse
Affiliation(s)
| | | | | | - Sarah E Lindo
- HHMI Janelia Research Campus, Ashburn, VA 20147, USA
| | | |
Collapse
|
288
|
Cell-Type-Specific Dynamics of Calcium Activity in Cortical Circuits over the Course of Slow-Wave Sleep and Rapid Eye Movement Sleep. J Neurosci 2021; 41:4212-4222. [PMID: 33833082 PMCID: PMC8143210 DOI: 10.1523/jneurosci.1957-20.2021] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 02/19/2021] [Accepted: 02/23/2021] [Indexed: 11/21/2022] Open
Abstract
Sleep shapes cortical network activity, fostering global homeostatic downregulation of excitability while maintaining or even upregulating excitability in selected networks in a manner that supports memory consolidation. Here, we used two-photon calcium imaging of cortical layer 2/3 neurons in sleeping male mice to examine how these seemingly opposing dynamics are balanced in cortical networks. During slow-wave sleep (SWS) episodes, mean calcium activity of excitatory pyramidal (Pyr) cells decreased. Simultaneously, however, variance in Pyr population calcium activity increased, contradicting the notion of a homogenous downregulation of network activity. Indeed, we identified a subpopulation of Pyr cells distinctly upregulating calcium activity during SWS, which were highly active during sleep spindles known to support mnemonic processing. Rapid eye movement (REM) episodes following SWS were associated with a general downregulation of Pyr cells, including the subpopulation of Pyr cells active during spindles, which persisted into following stages of sleep and wakefulness. Parvalbumin-positive inhibitory interneurons (PV-In) showed an increase in calcium activity during SWS episodes, while activity remained unchanged during REM sleep episodes. This supports the view that downregulation of Pyr calcium activity during SWS results from increased somatic inhibition via PV-In, whereas downregulation during REM sleep is achieved independently of such inhibitory activity. Overall, our findings show that SWS enables upregulation of select cortical circuits (likely those which were involved in mnemonic processing) through a spindle-related process, whereas REM sleep mediates general downregulation, possibly through synaptic re-normalization.SIGNIFICANCE STATEMENT Sleep is thought to globally downregulate cortical excitability and, concurrently, to upregulate synaptic connections in neuron ensembles with newly encoded memory, with upregulation representing a function of sleep spindles. Using in vivo two-photon calcium imaging in combination with surface EEG recordings, we classified cells based on their calcium activity during sleep spindles. Spindle-active pyramidal (Pyr) cells persistently increased calcium activity during slow-wave sleep (SWS) episodes while spindle-inactive cells decreased calcium activity. Subsequent rapid eye movement (REM) sleep episodes profoundly reduced calcium activity in both cell clusters. Results indicate that SWS allows for a spindle-related differential upregulation of ensembles whereas REM sleep functions to globally downregulate networks.
Collapse
|
289
|
Myristoylation alone is sufficient for PKA catalytic subunits to associate with the plasma membrane to regulate neuronal functions. Proc Natl Acad Sci U S A 2021; 118:2021658118. [PMID: 33876760 DOI: 10.1073/pnas.2021658118] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Myristoylation is a posttranslational modification that plays diverse functional roles in many protein species. The myristate moiety is considered insufficient for protein-membrane associations unless additional membrane-affinity motifs, such as a stretch of positively charged residues, are present. Here, we report that the electrically neutral N-terminal fragment of the protein kinase A catalytic subunit (PKA-C), in which myristoylation is the only functional motif, is sufficient for membrane association. This myristoylation can associate a fraction of PKA-C molecules or fluorescent proteins (FPs) to the plasma membrane in neuronal dendrites. The net neutral charge of the PKA-C N terminus is evolutionally conserved, even though its membrane affinity can be readily tuned by changing charges near the myristoylation site. The observed membrane association, while moderate, is sufficient to concentrate PKA activity at the membrane by nearly 20-fold and is required for PKA regulation of AMPA receptors at neuronal synapses. Our results indicate that myristoylation may be sufficient to drive functionally significant membrane association in the absence of canonical assisting motifs. This provides a revised conceptual base for the understanding of how myristoylation regulates protein functions.
Collapse
|
290
|
Assessing fatty acid-induced lipotoxicity and its therapeutic potential in glioblastoma using stimulated Raman microscopy. Sci Rep 2021; 11:7422. [PMID: 33795756 PMCID: PMC8016949 DOI: 10.1038/s41598-021-86789-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 03/16/2021] [Indexed: 01/15/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive primary brain tumor. The effectiveness of traditional therapies for GBM is limited and therefore new therapies are highly desired. Previous studies show that lipid metabolism reprogramming may be a potential therapeutic target in GBM. This study aims to evaluate the therapeutic potential of free fatty acid-induced lipotoxicity for the suppression of glioma growth. U87 glioma cells are treated with three fatty acids (FAs): palmitic acid (PA), oleic acid (OA), and eicosapentaenoic acid (EPA). Uptake of the FAs and formation of lipid droplets (LDs) are imaged and quantified using a lab-built stimulated Raman scattering (SRS) microscope. Our results show that a supply of 200 µM PA, OA, and EPA leads to efficient LDs accumulation in glioma cells. We find that inhibition of triglycerides (TAGs) synthesis depletes LDs and enhances lipotoxicity, which is evidenced by the reduced cell proliferation rates. In particular, our results suggest that EPA treatment combined with depletion of LDs significantly reduces the survival rate of glioma cells by more than 50%, indicating the therapeutic potential of this approach. Future work will focus on understanding the metabolic mechanism of EPA-induced lipotoxicity to further enhance its anticancer effects.
Collapse
|
291
|
Oh B, Wu Y, Swaminathan V, Lam V, Ding J, George PM. Modulating the Electrical and Mechanical Microenvironment to Guide Neuronal Stem Cell Differentiation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2002112. [PMID: 33854874 PMCID: PMC8025039 DOI: 10.1002/advs.202002112] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 12/08/2020] [Indexed: 05/27/2023]
Abstract
The application of induced pluripotent stem cells (iPSCs) in disease modeling and regenerative medicine can be limited by the prolonged times required for functional human neuronal differentiation and traditional 2D culture techniques. Here, a conductive graphene scaffold (CGS) to modulate mechanical and electrical signals to promote human iPSC-derived neurons is presented. The soft CGS with cortex-like stiffness (≈3 kPa) and electrical stimulation (±800 mV/100 Hz for 1 h) incurs a fivefold improvement in the rate (14d) of generating iPSC-derived neurons over some traditional protocols, with an increase in mature cellular markers and electrophysiological characteristics. Consistent with other culture conditions, it is found that the pro-neurogenic effects of mechanical and electrical stimuli rely on RhoA/ROCK signaling and de novo ciliary neurotrophic factor (CNTF) production respectively. Thus, the CGS system creates a combined physical and continuously modifiable, electrical niche to efficiently and quickly generate iPSC-derived neurons.
Collapse
Affiliation(s)
- Byeongtaek Oh
- Department of Neurology and Neurological SciencesStanford University School of MedicineStanfordCA94305USA
| | - Yu‐Wei Wu
- Department of NeurosurgeryStanford University School of MedicineStanfordCA94305USA
- Institute of Molecular BiologyAcademia SinicaTaiwan
| | - Vishal Swaminathan
- Department of Neurology and Neurological SciencesStanford University School of MedicineStanfordCA94305USA
| | - Vivek Lam
- Department of Neurology and Neurological SciencesStanford University School of MedicineStanfordCA94305USA
| | - Jun Ding
- Department of Neurology and Neurological SciencesStanford University School of MedicineStanfordCA94305USA
- Department of NeurosurgeryStanford University School of MedicineStanfordCA94305USA
| | - Paul M. George
- Department of Neurology and Neurological SciencesStanford University School of MedicineStanfordCA94305USA
| |
Collapse
|
292
|
Arlt C, Häusser M. Microcircuit Rules Governing Impact of Single Interneurons on Purkinje Cell Output In Vivo. Cell Rep 2021; 30:3020-3035.e3. [PMID: 32130904 PMCID: PMC7059114 DOI: 10.1016/j.celrep.2020.02.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 01/07/2020] [Accepted: 02/03/2020] [Indexed: 01/05/2023] Open
Abstract
The functional impact of single interneurons on neuronal output in vivo and how interneurons are recruited by physiological activity patterns remain poorly understood. In the cerebellar cortex, molecular layer interneurons and their targets, Purkinje cells, receive excitatory inputs from granule cells and climbing fibers. Using dual patch-clamp recordings from interneurons and Purkinje cells in vivo, we probe the spatiotemporal interactions between these circuit elements. We show that single interneuron spikes can potently inhibit Purkinje cell output, depending on interneuron location. Climbing fiber input activates many interneurons via glutamate spillover but results in inhibition of those interneurons that inhibit the same Purkinje cell receiving the climbing fiber input, forming a disinhibitory motif. These interneuron circuits are engaged during sensory processing, creating diverse pathway-specific response functions. These findings demonstrate how the powerful effect of single interneurons on Purkinje cell output can be sculpted by various interneuron circuit motifs to diversify cerebellar computations.
Collapse
Affiliation(s)
- Charlotte Arlt
- Wolfson Institute for Biomedical Research and Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| | - Michael Häusser
- Wolfson Institute for Biomedical Research and Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
293
|
Nojima T, Rings A, Allen AM, Otto N, Verschut TA, Billeter JC, Neville MC, Goodwin SF. A sex-specific switch between visual and olfactory inputs underlies adaptive sex differences in behavior. Curr Biol 2021; 31:1175-1191.e6. [PMID: 33508219 PMCID: PMC7987718 DOI: 10.1016/j.cub.2020.12.047] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/15/2020] [Accepted: 12/24/2020] [Indexed: 01/05/2023]
Abstract
Although males and females largely share the same genome and nervous system, they differ profoundly in reproductive investments and require distinct behavioral, morphological, and physiological adaptations. How can the nervous system, while bound by both developmental and biophysical constraints, produce these sex differences in behavior? Here, we uncover a novel dimorphism in Drosophila melanogaster that allows deployment of completely different behavioral repertoires in males and females with minimum changes to circuit architecture. Sexual differentiation of only a small number of higher order neurons in the brain leads to a change in connectivity related to the primary reproductive needs of both sexes-courtship pursuit in males and communal oviposition in females. This study explains how an apparently similar brain generates distinct behavioral repertoires in the two sexes and presents a fundamental principle of neural circuit organization that may be extended to other species.
Collapse
Affiliation(s)
- Tetsuya Nojima
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford OX1 3SR, UK
| | - Annika Rings
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford OX1 3SR, UK
| | - Aaron M Allen
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford OX1 3SR, UK
| | - Nils Otto
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford OX1 3SR, UK
| | - Thomas A Verschut
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, the Netherlands
| | - Jean-Christophe Billeter
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, the Netherlands
| | - Megan C Neville
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford OX1 3SR, UK.
| | - Stephen F Goodwin
- Centre for Neural Circuits and Behaviour, University of Oxford, Oxford OX1 3SR, UK.
| |
Collapse
|
294
|
Takahashi T, Herdzik KP, Bourdakos KN, Read JA, Mahajan S. Selective Imaging of Microplastic and Organic Particles in Flow by Multimodal Coherent Anti-Stokes Raman Scattering and Two-Photon Excited Autofluorescence Analysis. Anal Chem 2021; 93:5234-5240. [PMID: 33729769 DOI: 10.1021/acs.analchem.0c05474] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Microplastic pollution is an urgent global issue. While spectroscopic techniques have been widely used for the identification of plastics collected from aquatic environments, these techniques are often labor-intensive and time-consuming due to sample collection, preparation, and long measurement times. In this study, a method for the two-dimensional detection and classification of flowing microplastic and organic biotic particles with high spatial and temporal resolutions has been proposed based on the simultaneous detection of coherent anti-Stokes Raman scattering (CARS) and two-photon excited autofluorescence (TPEAF) signals. Poly(methyl methacrylate) (PMMA), polystyrene (PS), and low-density polyethylene (LDPE) particles with sizes ranging from several tens to hundreds of micrometers were selectively detected in flow with an average velocity of 4.17 mm/s by CARS line scanning. With the same flow velocity, flowing PMMA and alga particles were measured using a multimodal system of CARS and TPEAF signals. The average intensities of both PMMA and alga particles in the CARS signals at a frequency of 2940 cm-1 were higher than the background level, while only algae emitted TPEAF signals. This allowed the classification of PMMA and alga particles to be successfully performed in flow by the simultaneous detection of CARS and TPEAF signals. With the proposed method, the monitoring of microplastics in a continuous water flow without collection or extraction is possible, which is game-changing for the current sampling-based microplastic analysis.
Collapse
Affiliation(s)
- Tomoko Takahashi
- Advanced Science-Technology Research Program (ASTER), Japan Agency for Marine-Earth Science and Technology (JAMSTEC), 2-15 Natsushima-cho, Yokosuka, Kanagawa 2370061, Japan.,School of Chemistry and the Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton SO17 1BJ, United Kingdom.,Institute of Industrial Science, The University of Tokyo,4-6-1 Komaba, Meguro-ku, Tokyo 1538505, Japan
| | - Krzysztof Pawel Herdzik
- School of Chemistry and the Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton SO17 1BJ, United Kingdom
| | - Konstantinos Nikolaos Bourdakos
- School of Chemistry and the Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton SO17 1BJ, United Kingdom
| | - James Arthur Read
- School of Chemistry and the Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton SO17 1BJ, United Kingdom
| | - Sumeet Mahajan
- School of Chemistry and the Institute for Life Sciences, University of Southampton, Highfield Campus, Southampton SO17 1BJ, United Kingdom
| |
Collapse
|
295
|
A versatile deep learning architecture for classification and label-free prediction of hyperspectral images. NAT MACH INTELL 2021; 3:306-315. [PMID: 34676358 PMCID: PMC8528004 DOI: 10.1038/s42256-021-00309-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hyperspectral imaging is a technique that provides rich chemical or compositional information not regularly available to traditional imaging modalities such as intensity imaging or color imaging based on the reflection, transmission, or emission of light. Analysis of hyperspectral imaging often relies on machine learning methods to extract information. Here, we present a new flexible architecture, the U-within-U-Net, that can perform classification, segmentation, and prediction of orthogonal imaging modalities on a variety of hyperspectral imaging techniques. Specifically, we demonstrate feature segmentation and classification on the Indian Pines hyperspectral dataset and simultaneous location prediction of multiple drugs in mass spectrometry imaging of rat liver tissue. We further demonstrate label-free fluorescence image prediction from hyperspectral stimulated Raman scattering microscopy images. The applicability of the U-within-U-Net architecture on diverse datasets with widely varying input and output dimensions and data sources suggest that it has great potential in advancing the use of hyperspectral imaging across many different application areas ranging from remote sensing, to medical imaging, to microscopy.
Collapse
|
296
|
Perez-Alvarez A, Huhn F, Dürst CD, Franzelin A, Lamothe-Molina PJ, Oertner TG. Freeze-Frame Imaging of Dendritic Calcium Signals With TubuTag. Front Mol Neurosci 2021; 14:635820. [PMID: 33762909 PMCID: PMC7982875 DOI: 10.3389/fnmol.2021.635820] [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: 11/30/2020] [Accepted: 02/09/2021] [Indexed: 11/16/2022] Open
Abstract
The extensive dendritic arbor of neurons is thought to be actively involved in the processing of information. Dendrites contain a rich diversity of ligand- and voltage-activated ion channels as well as metabotropic receptors. In addition, they are capable of releasing calcium from intracellular stores. Under specific conditions, large neurons produce calcium spikes that are locally restricted to a dendritic section. To investigate calcium signaling in dendrites, we introduce TubuTag, a genetically encoded ratiometric calcium sensor anchored to the cytoskeleton. TubuTag integrates cytoplasmic calcium signals by irreversible photoconversion from green to red fluorescence when illuminated with violet light. We used a custom two-photon microscope with a large field of view to image pyramidal neurons in CA1 at subcellular resolution. Photoconversion was strongest in the most distal parts of the apical dendrite, suggesting a gradient in the amplitude of dendritic calcium signals. As the read-out of fluorescence can be performed several hours after photoconversion, TubuTag will help investigating dendritic signal integration and calcium homeostasis in large populations of neurons.
Collapse
Affiliation(s)
- Alberto Perez-Alvarez
- Institute for Synaptic Physiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Rapp OptoElectronic GmbH, Wedel, Germany
| | | | - Céline D Dürst
- Institute for Synaptic Physiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Rapp OptoElectronic GmbH, Wedel, Germany
| | - Andreas Franzelin
- Institute for Synaptic Physiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Paul J Lamothe-Molina
- Institute for Synaptic Physiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas G Oertner
- Institute for Synaptic Physiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
297
|
Fournel R, Hartveit E, Veruki ML. Differential Contribution of Gap Junctions to the Membrane Properties of ON- and OFF-Bipolar Cells of the Rat Retina. Cell Mol Neurobiol 2021; 41:229-245. [PMID: 32323153 PMCID: PMC7870642 DOI: 10.1007/s10571-020-00845-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/08/2020] [Indexed: 01/31/2023]
Abstract
Gap junctions are ubiquitous within the retina, but in general, it remains to be determined whether gap junction coupling between specific cell types is sufficiently strong to mediate functionally relevant coupling via electrical synapses. From ultrastructural, tracer coupling and immunolabeling studies, there is clear evidence for gap junctions between cone bipolar cells, but it is not known if these gap junctions function as electrical synapses. Here, using whole-cell voltage-clamp recording in rat (male and female) retinal slices, we investigated whether the gap junctions of bipolar cells make a measurable contribution to the membrane properties of these cells. We measured the input resistance (RN) of bipolar cells before and after applying meclofenamic acid (MFA) to block gap junctions. In the presence of MFA, RN of ON-cone bipolar cells displayed a clear increase, paralleled by block of the electrical coupling between these cells and AII amacrine cells in recordings of coupled cell pairs. For OFF-cone and rod bipolar cells, RN did not increase in the presence of MFA. The results for rod bipolar cells are consistent with the lack of gap junctions in these cells. However, for OFF-cone bipolar cells, our results suggest that the morphologically identified gap junctions between these cells do not support a junctional conductance that is sufficient to mediate effective electrical coupling. Instead, these junctions might play a role in chemical and/or metabolic coupling between subcellular compartments.
Collapse
Affiliation(s)
- Rémi Fournel
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009, Bergen, Norway
| | - Espen Hartveit
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009, Bergen, Norway.
| | - Margaret Lin Veruki
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009, Bergen, Norway.
| |
Collapse
|
298
|
Sharma A, Goring A, Johnson PB, Emery RJH, Hesse E, Boyde A, Olsen BR, Pitsillides AA, Oreffo ROC, Mahajan S, Clarkin CE. Multiscale molecular profiling of pathological bone resolves sexually dimorphic control of extracellular matrix composition. Dis Model Mech 2021; 14:dmm048116. [PMID: 33563616 PMCID: PMC7988766 DOI: 10.1242/dmm.048116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/21/2021] [Indexed: 11/28/2022] Open
Abstract
Collagen assembly during development is essential for successful matrix mineralisation, which determines bone quality and mechanocompetence. However, the biochemical and structural perturbations that drive pathological skeletal collagen configuration remain unclear. Deletion of vascular endothelial growth factor (VEGF; also known as VEGFA) in bone-forming osteoblasts (OBs) induces sex-specific alterations in extracellular matrix (ECM) conformation and mineralisation coupled to vascular changes, which are augmented in males. Whether this phenotypic dimorphism arises as a result of the divergent control of ECM composition and its subsequent arrangement is unknown and is the focus of this study. Herein, we used murine osteocalcin-specific Vegf knockout (OcnVEGFKO) and performed ex vivo multiscale analysis at the tibiofibular junction of both sexes. Label-free and non-destructive polarisation-resolved second-harmonic generation (p-SHG) microscopy revealed a reduction in collagen fibre number in males following the loss of VEGF, complemented by observable defects in matrix organisation by backscattered electron scanning electron microscopy. This was accompanied by localised divergence in collagen orientation, determined by p-SHG anisotropy measurements, as a result of OcnVEGFKO. Raman spectroscopy confirmed that the effect on collagen was linked to molecular dimorphic VEGF effects on collagen-specific proline and hydroxyproline, and collagen intra-strand stability, in addition to matrix carbonation and mineralisation. Vegf deletion in male and female murine OB cultures in vitro further highlighted divergence in genes regulating local ECM structure, including Adamts2, Spp1, Mmp9 and Lama1. Our results demonstrate the utility of macromolecular imaging and spectroscopic modalities for the detection of collagen arrangement and ECM composition in pathological bone. Linking the sex-specific genetic regulators to matrix signatures could be important for treatment of dimorphic bone disorders that clinically manifest in pathological nano- and macro-level disorganisation. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Aikta Sharma
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton SO17 1BJ, UK
| | - Alice Goring
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton SO17 1BJ, UK
| | - Peter B. Johnson
- School of Chemistry and Institute for Life Sciences, Highfield Campus, University of Southampton, Southampton SO17 1BJ, UK
| | - Roger J. H. Emery
- Department of Surgery and Cancer, Faculty of Medicine, St Mary's Campus, Imperial College London, London W2 1PG, UK
| | - Eric Hesse
- Institute of Molecular Musculoskeletal Research, Faculty of Medicine, LMU Munich, Planegg-Martinsried, Munich 80336, Germany
| | - Alan Boyde
- Dental Physical Sciences, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 4NS, UK
| | - Bjorn R. Olsen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Andrew A. Pitsillides
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London NW1 0TU, UK
| | - Richard O. C. Oreffo
- Centre for Human Development, Stem Cell and Regeneration, Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Sumeet Mahajan
- School of Chemistry and Institute for Life Sciences, Highfield Campus, University of Southampton, Southampton SO17 1BJ, UK
| | - Claire E. Clarkin
- School of Biological Sciences, Highfield Campus, University of Southampton, Southampton SO17 1BJ, UK
| |
Collapse
|
299
|
ZHANG KANGNING, HU JUNJIE, YANG WEIJIAN. Deep Compressed Imaging via Optimized-Pattern Scanning. PHOTONICS RESEARCH 2021; 9:B57-B70. [PMID: 34532505 PMCID: PMC8443127 DOI: 10.1364/prj.410556] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 01/13/2021] [Indexed: 05/31/2023]
Abstract
The need for high-speed imaging in applications such as biomedicine, surveillance and consumer electronics has called for new developments of imaging systems. While the industrial effort continuously pushes the advance of silicon focal plane array image sensors, imaging through a single-pixel detector has gained significant interests thanks to the development of computational algorithms. Here, we present a new imaging modality, Deep Compressed Imaging via Optimized-Pattern Scanning (DeCIOPS), which can significantly increase the acquisition speed for a single-detector-based imaging system. We project and scan an illumination pattern across the object and collect the sampling signal with a single-pixel detector. We develop an innovative end-to-end optimized auto-encoder, using a deep neural network and compressed sensing algorithm, to optimize the illumination pattern, which allows us to reconstruct faithfully the image from a small number of samples, and with a high frame rate. Compared with the conventional switching-mask based single-pixel camera and point scanning imaging systems, our method achieves a much higher imaging speed, while retaining a similar imaging quality. We experimentally validated this imaging modality in the settings of both continuous-wave (CW) illumination and pulsed light illumination and showed high-quality image reconstructions with a high compressed sampling rate. This new compressed sensing modality could be widely applied in different imaging systems, enabling new applications which require high imaging speed.
Collapse
Affiliation(s)
- KANGNING ZHANG
- Department of Electrical and Computer Engineering, University of California, Davis, CA 95616, USA
| | - JUNJIE HU
- Department of Electrical and Computer Engineering, University of California, Davis, CA 95616, USA
| | - WEIJIAN YANG
- Department of Electrical and Computer Engineering, University of California, Davis, CA 95616, USA
| |
Collapse
|
300
|
Díaz-García CM, Meyer DJ, Nathwani N, Rahman M, Martínez-François JR, Yellen G. The distinct roles of calcium in rapid control of neuronal glycolysis and the tricarboxylic acid cycle. eLife 2021; 10:e64821. [PMID: 33555254 PMCID: PMC7870136 DOI: 10.7554/elife.64821] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/26/2021] [Indexed: 12/31/2022] Open
Abstract
When neurons engage in intense periods of activity, the consequent increase in energy demand can be met by the coordinated activation of glycolysis, the tricarboxylic acid (TCA) cycle, and oxidative phosphorylation. However, the trigger for glycolytic activation is unknown and the role for Ca2+ in the mitochondrial responses has been debated. Using genetically encoded fluorescent biosensors and NAD(P)H autofluorescence imaging in acute hippocampal slices, here we find that Ca2+ uptake into the mitochondria is responsible for the buildup of mitochondrial NADH, probably through Ca2+ activation of dehydrogenases in the TCA cycle. In the cytosol, we do not observe a role for the Ca2+/calmodulin signaling pathway, or AMPK, in mediating the rise in glycolytic NADH in response to acute stimulation. Aerobic glycolysis in neurons is triggered mainly by the energy demand resulting from either Na+ or Ca2+ extrusion, and in mouse dentate granule cells, Ca2+ creates the majority of this demand.
Collapse
Affiliation(s)
| | - Dylan J Meyer
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Nidhi Nathwani
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | - Mahia Rahman
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| | | | - Gary Yellen
- Department of Neurobiology, Harvard Medical SchoolBostonUnited States
| |
Collapse
|