51
|
Yang JS, Jang HJ, Sung KW, Rhie DJ, Yoon SH. Roles of metabotropic glutamate receptor 5 in low [Mg 2+] o-induced interictal epileptiform activity in rat hippocampal slices. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2024; 28:413-422. [PMID: 39198222 PMCID: PMC11362004 DOI: 10.4196/kjpp.2024.28.5.413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/26/2024] [Accepted: 05/01/2024] [Indexed: 09/01/2024]
Abstract
Group I metabotropic glutamate receptors (mGluRs) modulate postsynaptic neuronal excitability and epileptogenesis. We investigated roles of group I mGluRs on low extracellular Mg2+ concentration ([Mg2+]o)-induced epileptiform activity and neuronal cell death in the CA1 regions of isolated rat hippocampal slices without the entorhinal cortex using extracellular recording and propidium iodide staining. Exposure to Mg2+-free artificial cerebrospinal fluid can induce interictal epileptiform activity in the CA1 regions of rat hippocampal slices. MPEP, a mGluR 5 antagonist, significantly inhibited the spike firing of the low [Mg2+]o-induced epileptiform activity, whereas LY367385, a mGluR1 antagonist, did not. DHPG, a group 1 mGluR agonist, significantly increased the spike firing of the epileptiform activity. U73122, a PLC inhibitor, inhibited the spike firing. Thapsigargin, an ER Ca2+-ATPase antagonist, significantly inhibited the spike firing and amplitude of the epileptiform activity. Both the IP3 receptor antagonist 2-APB and the ryanodine receptor antagonist dantrolene significantly inhibited the spike firing. The PKC inhibitors such as chelerythrine and GF109203X, significantly increased the spike firing. Flufenamic acid, a relatively specific TRPC 1, 4, 5 channel antagonist, significantly inhibited the spike firing, whereas SKF96365, a relatively non-specific TRPC channel antagonist, did not. MPEP significantly decreased low [Mg2+]o DMEM-induced neuronal cell death in the CA1 regions, but LY367385 did not. We suggest that mGluR 5 is involved in low [Mg2+]oinduced interictal epileptiform activity in the CA1 regions of rat hippocampal slices through PLC, release of Ca2+ from intracellular stores and PKC and TRPC channels, which could be involved in neuronal cell death.
Collapse
Affiliation(s)
- Ji Seon Yang
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Catholic Neuroscience Institute, The Catholic University of Korea, Seoul 06591, Korea
| | - Hyun-Jong Jang
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Catholic Neuroscience Institute, The Catholic University of Korea, Seoul 06591, Korea
| | - Ki-Wug Sung
- Catholic Neuroscience Institute, The Catholic University of Korea, Seoul 06591, Korea
- Department of Pharmacology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Duck-Joo Rhie
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Catholic Neuroscience Institute, The Catholic University of Korea, Seoul 06591, Korea
| | - Shin Hee Yoon
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Catholic Neuroscience Institute, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
52
|
Bak A, Schmied K, Jakob ML, Bedogni F, Squire OA, Gittel B, Jesinghausen M, Schünemann KD, Weber Y, Kampa B, van Loo KMJ, Koch H. Temporal dynamics of neocortical development in organotypic mouse brain cultures: a comprehensive analysis. J Neurophysiol 2024; 132:1038-1055. [PMID: 39140591 DOI: 10.1152/jn.00178.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024] Open
Abstract
Murine organotypic brain slice cultures have been widely used in neuroscientific research and are offering the opportunity to study neuronal function under normal and disease conditions. Despite the broad application, the mechanisms governing the maturation of immature cortical circuits in vitro are not well understood. In this study, we present a detailed investigation into the development of the neocortex in vitro. Using a holistic approach, we studied organotypic whole hemisphere brain slice cultures from postnatal mice and tracked the development of the somatosensory area over a 5-wk period. Our analysis revealed the maturation of passive and active intrinsic properties of pyramidal cells together with their morphology, closely resembling in vivo development. Detailed multielectrode array (MEA) electrophysiological assessments and RNA expression profiling demonstrated stable network properties by 2 wk in culture, followed by the transition of spontaneous activity toward more complex patterns including high-frequency oscillations. However, culturing weeks 4 and 5 exhibited increased variability and initial signs of neuronal loss, highlighting the importance of considering developmental stages in experimental design. This comprehensive characterization is vital for understanding the temporal dynamics of the neocortical development in vitro, with implications for neuroscientific research methodologies, particularly in the investigation of diseases such as epilepsy and other neurodevelopmental disorders.NEW & NOTEWORTHY The development of the mouse neocortex in vitro mimics the in vivo development. Mouse brain cultures can serve as a model system for cortical development for the first 2 wk in vitro and as a model system for the adult cortex from 2 to 4 wk in vitro. Mouse organotypic brain slice cultures develop high-frequency network oscillations at γ frequency after 2 wk in vitro. Mouse brain cultures exhibit increased heterogeneity and variability after 4 wk in culture.
Collapse
Affiliation(s)
- Aniella Bak
- Department of Epileptology, Neurology, RWTH Aachen University Hospital Aachen, Aachen, Germany
| | - Katharina Schmied
- Department of Epileptology, Neurology, RWTH Aachen University Hospital Aachen, Aachen, Germany
| | - Morten L Jakob
- Systems Neurophysiology, Institute of Zoology, RWTH Aachen University, Aachen, Germany
| | - Francesco Bedogni
- School of Medicine, Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Olivia A Squire
- School of Medicine, Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Birgit Gittel
- Department of Epileptology, Neurology, RWTH Aachen University Hospital Aachen, Aachen, Germany
| | - Maik Jesinghausen
- Systems Neurophysiology, Institute of Zoology, RWTH Aachen University, Aachen, Germany
| | - Kerstin D Schünemann
- Department of Epileptology, Neurology, RWTH Aachen University Hospital Aachen, Aachen, Germany
| | - Yvonne Weber
- Department of Epileptology, Neurology, RWTH Aachen University Hospital Aachen, Aachen, Germany
| | - Björn Kampa
- Systems Neurophysiology, Institute of Zoology, RWTH Aachen University, Aachen, Germany
- JARA BRAIN, Institute of Neuroscience and Medicine (INM-10), Forschungszentrum Jülich, Jülich, Germany
| | - Karen M J van Loo
- Department of Epileptology, Neurology, RWTH Aachen University Hospital Aachen, Aachen, Germany
- Department of Neurosurgery, RWTH Aachen University Hospital Aachen, Aachen, Germany
| | - Henner Koch
- Department of Epileptology, Neurology, RWTH Aachen University Hospital Aachen, Aachen, Germany
| |
Collapse
|
53
|
Zou J, McNair E, DeCastro S, Lyons SP, Mordant A, Herring LE, Vetreno RP, Coleman LG. Microglia either promote or restrain TRAIL-mediated excitotoxicity caused by Aβ 1-42 oligomers. J Neuroinflammation 2024; 21:215. [PMID: 39218898 PMCID: PMC11367981 DOI: 10.1186/s12974-024-03208-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) features progressive neurodegeneration and microglial activation that results in dementia and cognitive decline. The release of soluble amyloid (Aβ) oligomers into the extracellular space is an early feature of AD pathology. This can promote excitotoxicity and microglial activation. Microglia can adopt several activation states with various functional outcomes. Protective microglial activation states have been identified in response to Aβ plaque pathology in vivo. However, the role of microglia and immune mediators in neurotoxicity induced by soluble Aβ oligomers is unclear. Further, there remains a need to identify druggable molecular targets that promote protective microglial states to slow or prevent the progression of AD. METHODS Hippocampal entorhinal brain slice culture (HEBSC) was employed to study mechanisms of Aβ1-42 oligomer-induced neurotoxicity as well as the role of microglia. The roles of glutamate hyperexcitation and immune signaling in Aβ-induced neurotoxicity were assessed using MK801 and neutralizing antibodies to the TNF-related apoptosis-inducing ligand (TRAIL) respectively. Microglial activation state was manipulated using Gi-hM4di designer receptor exclusively activated by designer drugs (DREADDs), microglial depletion with the colony-stimulating factor 1 receptor (CSF1R) antagonist PLX3397, and microglial repopulation (PLX3397 withdrawal). Proteomic changes were assessed by LC-MS/MS in microglia isolated from control, repopulated, or Aβ-treated HEBSCs. RESULTS Neurotoxicity induced by soluble Aβ1-42 oligomers involves glutamatergic hyperexcitation caused by the proinflammatory mediator and death receptor ligand TRAIL. Microglia were found to have the ability to both promote and restrain Aβ-induced toxicity. Induction of microglial Gi-signaling with hM4di to prevent pro-inflammatory activation blunted Aβ neurotoxicity, while microglial depletion with CSF1R antagonism worsened neurotoxicity caused by Aβ as well as TRAIL. HEBSCs with repopulated microglia, however, showed a near complete resistance to Aβ-induced neurotoxicity. Comparison of microglial proteomes revealed that repopulated microglia have a baseline anti-inflammatory and trophic phenotype with a predicted pathway activation that is nearly opposite that of Aβ-exposed microglia. mTORC2 and IRF7 were identified as potential targets for intervention. CONCLUSION Microglia are key mediators of both protection and neurodegeneration in response to Aβ. Polarizing microglia toward a protective state could be used as a preventative strategy against Aβ-induced neurotoxicity.
Collapse
Affiliation(s)
- Jian Zou
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, 27599, USA
| | - Elizabeth McNair
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, 27599, USA
| | - Sagan DeCastro
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, 27599, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, 27599, USA
| | - Scott P Lyons
- Department of Pharmacology, UNC Proteomics Core, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, 27599, USA
| | - Angie Mordant
- Department of Pharmacology, UNC Proteomics Core, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, 27599, USA
| | - Laura E Herring
- Department of Pharmacology, UNC Proteomics Core, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, 27599, USA
| | - Ryan P Vetreno
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, 27599, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, 27599, USA
| | - Leon G Coleman
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, 27599, USA.
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
54
|
Charmarke-Askar I, Spenlé C, Bagnard D. Complementary strategies to be used in conjunction with animal models for multiple sclerosis drug discovery: adapting preclinical validation of drug candidates to the need of remyelinating strategies. Expert Opin Drug Discov 2024; 19:1115-1124. [PMID: 39039755 DOI: 10.1080/17460441.2024.2382180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 07/16/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION The quest for novel MS therapies focuses on promoting remyelination and neuroprotection, necessitating innovative drug design paradigms and robust preclinical validation methods to ensure efficient clinical translation. The complexity of new drugs action mechanisms is strengthening the need for solid biological validation attempting to address all possible pitfalls and biases precluding access to efficient and safe drugs. AREAS COVERED In this review, the authors describe the different in vitro and in vivo models that should be used to create an integrated approach for preclinical validation of novel drugs, including the evaluation of the action mechanism. This encompasses 2D, 3D in vitro models and animal models presented in such a way to define the appropriate use in a global process of drug screening and hit validation. EXPERT OPINION None of the current available tests allow the concomitant evaluation of anti-inflammatory, immune regulators or remyelinating agents with sufficient reliability. Consequently, the collaborative efforts of academia, industry, and regulatory agencies are essential for establishing standardized protocols, validating novel methodologies, and translating preclinical findings into clinically meaningful outcomes.
Collapse
|
55
|
Chater TE, Eggl MF, Goda Y, Tchumatchenko T. Competitive processes shape multi-synapse plasticity along dendritic segments. Nat Commun 2024; 15:7572. [PMID: 39217140 PMCID: PMC11365941 DOI: 10.1038/s41467-024-51919-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
Neurons receive thousands of inputs onto their dendritic arbour, where individual synapses undergo activity-dependent plasticity. Long-lasting changes in postsynaptic strengths correlate with changes in spine head volume. The magnitude and direction of such structural plasticity - potentiation (sLTP) and depression (sLTD) - depend upon the number and spatial distribution of stimulated synapses. However, how neurons allocate resources to implement synaptic strength changes across space and time amongst neighbouring synapses remains unclear. Here we combined experimental and modelling approaches to explore the elementary processes underlying multi-spine plasticity. We used glutamate uncaging to induce sLTP at varying number of synapses sharing the same dendritic branch, and we built a model incorporating a dual role Ca2+-dependent component that induces spine growth or shrinkage. Our results suggest that competition among spines for molecular resources is a key driver of multi-spine plasticity and that spatial distance between simultaneously stimulated spines impacts the resulting spine dynamics.
Collapse
Affiliation(s)
- Thomas E Chater
- Laboratory for Synaptic Plasticity and Connectivity, RIKEN Center for Brain Science, Wako-shi, Saitama, Japan
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Maximilian F Eggl
- Institute of Experimental Epileptology and Cognition Research, University of Bonn Medical Center, Venusberg-Campus 1, 53127, Bonn, Germany
- Institute of Neuroscience, CSIC-UMH, Alicante, Spain
| | - Yukiko Goda
- Laboratory for Synaptic Plasticity and Connectivity, RIKEN Center for Brain Science, Wako-shi, Saitama, Japan.
- Synapse Biology Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Kunigami-gun, Okinawa, Japan.
| | - Tatjana Tchumatchenko
- Institute of Experimental Epileptology and Cognition Research, University of Bonn Medical Center, Venusberg-Campus 1, 53127, Bonn, Germany.
| |
Collapse
|
56
|
Brandhorst E, Xu L, Klimezak M, Goegan B, Hong H, Hammes HP, Specht A, Cambridge S. In Vivo Optogenetic Manipulation of Transgene Expression in Retinal Neurovasculature. JACS AU 2024; 4:2818-2825. [PMID: 39211617 PMCID: PMC11350597 DOI: 10.1021/jacsau.4c00434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 05/27/2024] [Accepted: 06/14/2024] [Indexed: 09/04/2024]
Abstract
The retina is prone to developing pathological neovascularization, a leading cause of blindness in humans. Because excess neovascularization does not affect the entire retina, global inhibition treatment of angiogenesis critically interferes with healthy, unaffected retinal tissue. We therefore established an in vivo photoactivated gene expression paradigm which would allow light-mediated targeting of antiangiogenic genetic treatment only to affected retinal regions. We synthesized a "caged" (i.e., reversibly inhibited) photosensitive 4-hydroxytamoxifen analog. Molecular docking analyses validated its reduced transcriptional activity. Caged 4-hydroxytamoxifen was intravitreally injected into mice harboring the inducible Cre/lox system, with CreERT2 being expressed via the Tie2 promoter in the neurovasculature. Subsequent in vivo irradiation of eyes significantly induced retinal expression of a Cre-dependent transgene in retinal blood vessels. Using GFAP-CreERT2 mice, successful photoactivation was also achieved in eyes and also in ex vivo brain slices for validation of the approach. This highlights the possibility of light-mediated gene therapies specific for the retina, a key first step in personalized medicine.
Collapse
Affiliation(s)
- Eric Brandhorst
- Fifth Medical Department, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Liang Xu
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, Arkansas 72079, United States
| | - Maxime Klimezak
- Laboratoire de Chémo-Biologie Synthétique et Thérapeutique, Equipe Nanoparticule Intelligentes, Université de Strasbourg, CNRS, CBST UMR 7199, F-67000 Strasbourg, France
| | - Bastien Goegan
- Laboratoire de Chémo-Biologie Synthétique et Thérapeutique, Equipe Nanoparticule Intelligentes, Université de Strasbourg, CNRS, CBST UMR 7199, F-67000 Strasbourg, France
| | - Huixiao Hong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, Arkansas 72079, United States
| | - Hans-Peter Hammes
- Fifth Medical Department, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Alexandre Specht
- Laboratoire de Chémo-Biologie Synthétique et Thérapeutique, Equipe Nanoparticule Intelligentes, Université de Strasbourg, CNRS, CBST UMR 7199, F-67000 Strasbourg, France
| | - Sidney Cambridge
- Dr. Senckenberg Anatomy, Anatomy II, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany
- Department of Medicine, Health and Medical University Potsdam, 14471 Potsdam, Germany
| |
Collapse
|
57
|
Xiong WH, Qin M, Zhong H. PKA regulation of neuronal function requires the dissociation of catalytic subunits from regulatory subunits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.05.556437. [PMID: 37732264 PMCID: PMC10508765 DOI: 10.1101/2023.09.05.556437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Protein kinase A (PKA) plays essential roles in diverse cellular functions. However, the spatiotemporal dynamics of endogenous PKA upon activation remain debated. The classical model predicts that PKA catalytic subunits dissociate from regulatory subunits in the presence of cAMP, whereas a second model proposes that catalytic subunits remain associated with regulatory subunits following physiological activation. Here we report that different PKA subtypes, as defined by the regulatory subunit, exhibit distinct subcellular localization at rest in CA1 neurons of cultured hippocampal slices. Nevertheless, when all tested PKA subtypes are activated by norepinephrine, presumably via the β-adrenergic receptor, catalytic subunits translocate to dendritic spines but regulatory subunits remain unmoved. These differential spatial dynamics between the subunits indicate that at least a significant fraction of PKA dissociates. Furthermore, PKA-dependent regulation of synaptic plasticity and transmission can be supported only by wildtype, dissociable PKA, but not by inseparable PKA. These results indicate that endogenous PKA regulatory and catalytic subunits dissociate to achieve PKA function in neurons.
Collapse
Affiliation(s)
- Wei-Hong Xiong
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Maozhen Qin
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Haining Zhong
- Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
58
|
Rivers-Auty J, Hoyle C, Pointer A, Lawrence C, Pickering-Brown S, Brough D, Ryan S. C9orf72 dipeptides activate the NLRP3 inflammasome. Brain Commun 2024; 6:fcae282. [PMID: 39229486 PMCID: PMC11369816 DOI: 10.1093/braincomms/fcae282] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/24/2024] [Accepted: 08/16/2024] [Indexed: 09/05/2024] Open
Abstract
Frontotemporal dementia and amyotrophic lateral sclerosis are neurodegenerative diseases with considerable clinical, genetic and pathological overlap. The most common cause of both diseases is a hexanucleotide repeat expansion in C9orf72. The expansion is translated to produce five toxic dipeptides, which aggregate in patient brain. Neuroinflammation is a feature of frontotemporal dementia and amyotrophic lateral sclerosis; however, its causes are unknown. The nod-like receptor family, pyrin domain-containing 3 inflammasome is implicated in several other neurodegenerative diseases as a driver of damaging inflammation. The inflammasome is a multi-protein complex which forms in immune cells in response to tissue damage, pathogens or aggregating proteins. Inflammasome activation is observed in models of other neurodegenerative diseases such as Alzheimer's disease, and inflammasome inhibition rescues cognitive decline in rodent models of Alzheimer's disease. Here, we show that a dipeptide arising from the C9orf72 expansion, poly-glycine-arginine, activated the inflammasome in microglia and macrophages, leading to secretion of the pro-inflammatory cytokine, interleukin-1β. Poly-glycine-arginine also activated the inflammasome in organotypic hippocampal slice cultures, and immunofluorescence imaging demonstrated formation of inflammasome specks in response to poly-glycine-arginine. Several clinically available anti-inflammatory drugs rescued poly-glycine-arginine-induced inflammasome activation. These data suggest that C9orf72 dipeptides contribute to the neuroinflammation observed in patients, and highlight the inflammasome as a potential therapeutic target for frontotemporal dementia and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Jack Rivers-Auty
- School of Medicine, University of Tasmania, Hobart, TAS 7000, Australia
| | - Christopher Hoyle
- Division of Neuroscience, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester M13 9PL, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester M13 9PT, UK
| | - Ayesha Pointer
- Division of Neuroscience, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester M13 9PL, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester M13 9PT, UK
| | - Catherine Lawrence
- Division of Neuroscience, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester M13 9PL, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester M13 9PT, UK
| | - Stuart Pickering-Brown
- Division of Neuroscience, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester M13 9PL, UK
| | - David Brough
- Division of Neuroscience, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester M13 9PL, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester M13 9PT, UK
| | - Sarah Ryan
- Division of Neuroscience, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester M13 9PL, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
59
|
Lamba K, Rani S. A novel approach of brain-computer interfacing (BCI) and Grad-CAM based explainable artificial intelligence: Use case scenario for smart healthcare. J Neurosci Methods 2024; 408:110159. [PMID: 38723868 DOI: 10.1016/j.jneumeth.2024.110159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/02/2024] [Accepted: 04/29/2024] [Indexed: 05/19/2024]
Abstract
BACKGROUND In order to push the frontiers of brain-computer interface (BCI) and neuron-electronics, this research presents a novel framework that combines cutting-edge technologies for improved brain-related diagnostics in smart healthcare. This research offers a ground-breaking application of transparent strategies to BCI, promoting openness and confidence in brain-computer interactions and taking inspiration from Grad-CAM (Gradient-weighted Class Activation Mapping) based Explainable Artificial Intelligence (XAI) methodology. The landscape of healthcare diagnostics is about to be redefined by the integration of various technologies, especially when it comes to illnesses related to the brain. NEW METHOD A novel approach has been proposed in this study comprising of Xception architecture which is trained on imagenet database following transfer learning process for extraction of significant features from magnetic resonance imaging dataset acquired from publicly available distinct sources as an input and linear support vector machine has been used for distinguishing distinct classes.Afterwards, gradient-weighted class activation mapping has been deployed as the foundation for explainable artificial intelligence (XAI) for generating informative heatmaps, representing spatial localization of features which were focused to achieve model's predictions. RESULTS Thus, the proposed model not only provides accurate outcomes but also provides transparency for the predictions generated by the Xception network to diagnose presence of abnormal tissues and avoids overfitting issues. Hyperparameters along with performance-metrics are also obtained while validating the proposed network on unseen brain MRI scans to ensure effectiveness of the proposed network. COMPARISON WITH EXISTING METHODS AND CONCLUSIONS The integration of Grad-CAM based explainable artificial intelligence with deep neural network namely Xception offers a significant impact in diagnosing brain tumor disease while highlighting the specific regions of input brain MRI images responsible for making predictions. In this study, the proposed network results in 98.92% accuracy, 98.15% precision, 99.09% sensitivity, 98.18% specificity and 98.91% dice-coefficient while identifying presence of abnormal tissues in the brain. Thus, Xception model trained on distinct dataset following transfer learning process offers remarkable diagnostic accuracy and linear support vector act as a classifier to provide efficient classification among distinct classes. In addition, the deployed explainable artificial intelligence approach helps in revealing the reasoning behind predictions made by deep neural network having black-box nature and provides a clear perspective to assist medical experts in achieving trustworthiness and transparency while diagnosing brain tumor disease in the smart healthcare.
Collapse
Affiliation(s)
- Kamini Lamba
- Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab 140401, India.
| | - Shalli Rani
- Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab 140401, India.
| |
Collapse
|
60
|
Vincent PFY, Young ED, Edge ASB, Glowatzki E. Auditory hair cells and spiral ganglion neurons regenerate synapses with refined release properties in vitro. Proc Natl Acad Sci U S A 2024; 121:e2315599121. [PMID: 39058581 PMCID: PMC11294990 DOI: 10.1073/pnas.2315599121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 06/12/2024] [Indexed: 07/28/2024] Open
Abstract
Ribbon synapses between inner hair cells (IHCs) and type I spiral ganglion neurons (SGNs) in the inner ear are damaged by noise trauma and with aging, causing "synaptopathy" and hearing loss. Cocultures of neonatal denervated organs of Corti and newly introduced SGNs have been developed to find strategies for improving IHC synapse regeneration, but evidence of the physiological normality of regenerated synapses is missing. This study utilizes IHC optogenetic stimulation and SGN recordings, showing that, when P3-5 denervated organs of Corti are cocultured with SGNs, newly formed IHC/SGN synapses are indeed functional, exhibiting glutamatergic excitatory postsynaptic currents. When using older organs of Corti at P10-11, synaptic activity probed by deconvolution showed more mature release properties, closer to the specialized mode of IHC synaptic transmission crucial for coding the sound signal. This functional assessment of newly formed IHC synapses developed here, provides a powerful tool for testing approaches to improve synapse regeneration.
Collapse
Affiliation(s)
- Philippe F. Y. Vincent
- The Center for Hearing and Balance, The Johns Hopkins School of Medicine, Baltimore, MD21205
- Department of Otolaryngology Head and Neck Surgery, The Johns Hopkins School of Medicine, Baltimore, MD21205
| | - Eric D. Young
- The Center for Hearing and Balance, The Johns Hopkins School of Medicine, Baltimore, MD21205
- Department of Otolaryngology Head and Neck Surgery, The Johns Hopkins School of Medicine, Baltimore, MD21205
- Department of Neuroscience, The Johns Hopkins School of Medicine, Baltimore, MD21205
- Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, MD21205
| | - Albert S. B. Edge
- Department of Otolaryngology, Harvard Medical School, Boston, MA02115
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, MA02114
- Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, MA02115
- Harvard Stem Cell Institute, Cambridge, MA02139
| | - Elisabeth Glowatzki
- The Center for Hearing and Balance, The Johns Hopkins School of Medicine, Baltimore, MD21205
- Department of Otolaryngology Head and Neck Surgery, The Johns Hopkins School of Medicine, Baltimore, MD21205
- Department of Neuroscience, The Johns Hopkins School of Medicine, Baltimore, MD21205
| |
Collapse
|
61
|
Anjum R, Clarke VRJ, Nagasawa Y, Murakoshi H, Paradis S. Rem2 interacts with CaMKII at synapses and restricts long-term potentiation in hippocampus. PLoS One 2024; 19:e0301063. [PMID: 38995900 PMCID: PMC11244776 DOI: 10.1371/journal.pone.0301063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 06/11/2024] [Indexed: 07/14/2024] Open
Abstract
Synaptic plasticity, the process whereby neuronal connections are either strengthened or weakened in response to stereotyped forms of stimulation, is widely believed to represent the molecular mechanism that underlies learning and memory. The holoenzyme calcium/calmodulin-dependent protein kinase II (CaMKII) plays a well-established and critical role in the induction of a variety of forms of synaptic plasticity such as long-term potentiation (LTP), long-term depression (LTD) and depotentiation. Previously, we identified the GTPase Rem2 as a potent, endogenous inhibitor of CaMKII. Here, we report that knock out of Rem2 enhances LTP at the Schaffer collateral to CA1 synapse in hippocampus, consistent with an inhibitory action of Rem2 on CaMKII in vivo. Further, re-expression of WT Rem2 rescues the enhanced LTP observed in slices obtained from Rem2 conditional knock out (cKO) mice, while expression of a mutant Rem2 construct that is unable to inhibit CaMKII in vitro fails to rescue increased LTP. In addition, we demonstrate that CaMKII and Rem2 interact in dendritic spines using a 2pFLIM-FRET approach. Taken together, our data lead us to propose that Rem2 serves as a brake on synaptic potentiation via inhibition of CaMKII activity. Further, the enhanced LTP phenotype we observe in Rem2 cKO slices reveals a previously unknown role for Rem2 in the negative regulation of CaMKII function.
Collapse
Affiliation(s)
- Rabia Anjum
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, Massachusetts, United States of America
| | - Vernon R. J. Clarke
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Yutaro Nagasawa
- Department of Physiological Sciences, The Graduate University for Advanced Studies; Hayama, Kanagawa, Japan
- Supportive Center for Brain Research, National Institute for Physiological Sciences; Okazaki, Aichi, Japan
| | - Hideji Murakoshi
- Department of Physiological Sciences, The Graduate University for Advanced Studies; Hayama, Kanagawa, Japan
- Supportive Center for Brain Research, National Institute for Physiological Sciences; Okazaki, Aichi, Japan
| | - Suzanne Paradis
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, Massachusetts, United States of America
| |
Collapse
|
62
|
Chakrabarty A, Newey SE, Promi MM, Agbetiameh BK, Munro D, Brodersen PJN, Gothard G, Mahfooz K, Mengual JP, Vyazovskiy VV, Akerman CJ. sUPRa is a dual-color reporter for unbiased quantification of the unfolded protein response with cellular resolution. Sci Rep 2024; 14:14990. [PMID: 38951511 PMCID: PMC11217371 DOI: 10.1038/s41598-024-65611-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 06/21/2024] [Indexed: 07/03/2024] Open
Abstract
The unfolded protein response (UPR) maintains proteostasis upon endoplasmic reticulum (ER) stress, and is initiated by a range of physiological and pathological processes. While there have been advances in developing fluorescent reporters for monitoring individual signaling pathways of the UPR, this approach may not capture a cell's overall UPR activity. Here we describe a novel sensor of UPR activity, sUPRa, which is designed to report the global UPR. sUPRa displays excellent response characteristics, outperforms reporters of individual UPR pathways in terms of sensitivity and kinetics, and responds to a range of different ER stress stimuli. Furthermore, sUPRa's dual promoter and fluorescent protein design ensures that both UPR-active and inactive cells are detected, and controls for reporter copy number. Using sUPRa, we reveal UPR activation in layer 2/3 pyramidal neurons of mouse cerebral cortex following a period of sleep deprivation. sUPRa affords new opportunities for quantifying physiological UPR activity with cellular resolution.
Collapse
Affiliation(s)
- Atreyi Chakrabarty
- Department of Pharmacology, University of Oxford, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Sarah E Newey
- Department of Pharmacology, University of Oxford, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Maisha M Promi
- Department of Pharmacology, University of Oxford, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Belinda K Agbetiameh
- Department of Pharmacology, University of Oxford, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Daniella Munro
- Department of Pharmacology, University of Oxford, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Paul J N Brodersen
- Department of Pharmacology, University of Oxford, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Gemma Gothard
- Department of Pharmacology, University of Oxford, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Kashif Mahfooz
- Department of Pharmacology, University of Oxford, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Jose P Mengual
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Sherrington Road, Oxford, OX1 3PT, UK
| | - Vladyslav V Vyazovskiy
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Sherrington Road, Oxford, OX1 3PT, UK
| | - Colin J Akerman
- Department of Pharmacology, University of Oxford, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK.
| |
Collapse
|
63
|
Rao S, Liang F, Herring BE. RhoGEF Tiam2 Regulates Glutamatergic Synaptic Transmission in Hippocampal CA1 Pyramidal Neurons. eNeuro 2024; 11:ENEURO.0500-21.2024. [PMID: 38871458 PMCID: PMC11262554 DOI: 10.1523/eneuro.0500-21.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/29/2024] [Accepted: 05/23/2024] [Indexed: 06/15/2024] Open
Abstract
Glutamatergic synapses exhibit significant molecular diversity, but circuit-specific mechanisms that underlie synaptic regulation are not well characterized. Prior reports show that Rho-guanine nucleotide exchange factor (RhoGEF) Tiam1 regulates perforant path→dentate gyrus granule neuron synapses. In the present study, we report Tiam1's homolog Tiam2 is implicated in glutamatergic neurotransmission in CA1 pyramidal neurons. We find that Tiam2 regulates evoked excitatory glutamatergic currents via a postsynaptic mechanism mediated by the catalytic Dbl-homology domain. Overall, we present evidence for RhoGEF Tiam2's role in glutamatergic synapse function at Schaffer collateral→CA1 pyramidal neuron synapses.
Collapse
Affiliation(s)
- Sadhna Rao
- Department of Biological Sciences, Neurobiology Section, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California 90089
| | - Feng Liang
- Department of Biological Sciences, Neurobiology Section, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California 90089
| | - Bruce E Herring
- Department of Biological Sciences, Neurobiology Section, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, California 90089
| |
Collapse
|
64
|
Michalska JM, Lyudchik J, Velicky P, Štefaničková H, Watson JF, Cenameri A, Sommer C, Amberg N, Venturino A, Roessler K, Czech T, Höftberger R, Siegert S, Novarino G, Jonas P, Danzl JG. Imaging brain tissue architecture across millimeter to nanometer scales. Nat Biotechnol 2024; 42:1051-1064. [PMID: 37653226 PMCID: PMC11252008 DOI: 10.1038/s41587-023-01911-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 07/20/2023] [Indexed: 09/02/2023]
Abstract
Mapping the complex and dense arrangement of cells and their connectivity in brain tissue demands nanoscale spatial resolution imaging. Super-resolution optical microscopy excels at visualizing specific molecules and individual cells but fails to provide tissue context. Here we developed Comprehensive Analysis of Tissues across Scales (CATS), a technology to densely map brain tissue architecture from millimeter regional to nanometer synaptic scales in diverse chemically fixed brain preparations, including rodent and human. CATS uses fixation-compatible extracellular labeling and optical imaging, including stimulated emission depletion or expansion microscopy, to comprehensively delineate cellular structures. It enables three-dimensional reconstruction of single synapses and mapping of synaptic connectivity by identification and analysis of putative synaptic cleft regions. Applying CATS to the mouse hippocampal mossy fiber circuitry, we reconstructed and quantified the synaptic input and output structure of identified neurons. We furthermore demonstrate applicability to clinically derived human tissue samples, including formalin-fixed paraffin-embedded routine diagnostic specimens, for visualizing the cellular architecture of brain tissue in health and disease.
Collapse
Affiliation(s)
- Julia M Michalska
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Julia Lyudchik
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Philipp Velicky
- Institute of Science and Technology Austria, Klosterneuburg, Austria
- Core Facility Imaging, Medical University of Vienna, Vienna, Austria
| | - Hana Štefaničková
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Jake F Watson
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Alban Cenameri
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Christoph Sommer
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Nicole Amberg
- Department of Neurology, Division of Neuropathology and Neurochemistry, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | | | - Karl Roessler
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Thomas Czech
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Romana Höftberger
- Department of Neurology, Division of Neuropathology and Neurochemistry, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Sandra Siegert
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Gaia Novarino
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Peter Jonas
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Johann G Danzl
- Institute of Science and Technology Austria, Klosterneuburg, Austria.
| |
Collapse
|
65
|
Hawker B, Dhakal M, Connor B, McCaughey-Chapman A. Modeling demyelination and endogenous remyelination in spinal cord ex vivo rat organotypic slice cultures. Front Cell Neurosci 2024; 18:1345042. [PMID: 38988661 PMCID: PMC11233765 DOI: 10.3389/fncel.2024.1345042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 06/17/2024] [Indexed: 07/12/2024] Open
Abstract
Introduction Demyelination of the spinal cord is a prominent feature of multiple sclerosis (MS) and spinal cord injuries (SCI), where impaired neuronal communication between the brain and periphery has devastating consequences on neurological function. Demyelination precedes remyelination, an endogenous process in which oligodendrocyte precursor cells (OPCs) differentiate into mature, myelinating oligodendrocytes with the ability to restore the myelin sheath and reinstate functional nerve signaling. However, in MS or SCI, demyelination is more severe, persistent, and inhibitory to OPC-mediated remyelination, leading to a permanent loss of neuronal function. Currently, there are no effective treatments for demyelination, and existing pre-clinical models typically focus on brain tissue with little characterization of demyelination within the spinal cord. Organotypic slice cultures are a useful tool to study neurological disease, providing a more complex 3-dimensional system than standard 2-dimensional in vitro cell cultures. Methods Building on our previously developed rat brain slice culture protocol, we have extended our findings to develop a rat longitudinal spinal cord ex vivo model of demyelination. Results We generated rat longitudinal spinal cord slice cultures that remain viable for up to 6 weeks in culture and retain key anatomical features of the spinal cord's cytoarchitecture. We show that treating longitudinal spinal cord slices with lysolecithin (LPC) induced robust demyelination with some endogenous remyelination, which was not seen following exposure to lipopolysaccharide (LPS). Discussion Our ex vivo organotypic spinal cord slice culture system provides a platform to model demyelination and endogenous remyelination long-term, mimicking that observed in LPC-induced rodent models of demyelination. This platform is suitable for the development and testing of novel therapeutic strategies with ease of manipulation prior to in vivo experimentation.
Collapse
Affiliation(s)
| | | | | | - Amy McCaughey-Chapman
- Department of Pharmacology and Clinical Pharmacology, Centre for Brain Research, School of Medical Science, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
66
|
Prinkey K, Thompson E, Saikia J, Cid T, Dore K. Fluorescence lifetime imaging of AMPA receptor endocytosis in living neurons: effects of Aβ and PP1. Front Mol Neurosci 2024; 17:1409401. [PMID: 38915938 PMCID: PMC11194458 DOI: 10.3389/fnmol.2024.1409401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/22/2024] [Indexed: 06/26/2024] Open
Abstract
The relative amount of AMPA receptors expressed at the surface of neurons can be measured using superecliptic pHluorin (SEP) labeling at their N-terminus. However, the high signal variability resulting from protein overexpression in neurons and the low signal observed in intracellular vesicles make quantitative characterization of receptor trafficking difficult. Here, we establish a real-time live-cell assay of AMPAR trafficking based on fluorescence lifetime imaging (FLIM), which allows for simultaneous visualization of both surface and intracellular receptors. Using this assay, we found that elevating amyloid-beta (Aβ) levels leads to a strong increase in intracellular GluA1 and GluA2-containing receptors, indicating that Aβ triggers the endocytosis of these AMPARs. In APP/PS1 Alzheimer's disease model mouse neurons, FLIM revealed strikingly different AMPAR trafficking properties for GluA1- and GluA3-containing receptors, suggesting that chronic Aβ exposure triggered the loss of both surface and intracellular GluA3-containing receptors. Interestingly, overexpression of protein phosphatase 1 (PP1) also resulted in GluA1 endocytosis as well as depressed synaptic transmission, confirming the important role of phosphorylation in regulating AMPAR trafficking. This new approach allows for the quantitative measurement of extracellular pH, small changes in receptor trafficking, as well as simultaneous measurement of surface and internalized AMPARs in living neurons, and could therefore be applied to several different studies in the future.
Collapse
Affiliation(s)
| | | | | | | | - Kim Dore
- Center for Neural Circuits and Behavior, Department of Neuroscience, School of Medicine, University of California at San Diego, La Jolla, CA, United States
| |
Collapse
|
67
|
Barnett AM, Dawkins L, Zou J, McNair E, Nikolova VD, Moy SS, Sutherland GT, Stevens J, Colie M, Katemboh K, Kellner H, Damian C, DeCastro S, Vetreno RP, Coleman LG. Loss of neuronal lysosomal acid lipase drives amyloid pathology in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.09.596693. [PMID: 38915509 PMCID: PMC11195138 DOI: 10.1101/2024.06.09.596693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Underlying drivers of late-onset Alzheimer's disease (LOAD) pathology remain unknown. However, multiple biologically diverse risk factors share a common pathological progression. To identify convergent molecular abnormalities that drive LOAD pathogenesis we compared two common midlife risk factors for LOAD, heavy alcohol use and obesity. This revealed that disrupted lipophagy is an underlying cause of LOAD pathogenesis. Both exposures reduced lysosomal flux, with a loss of neuronal lysosomal acid lipase (LAL). This resulted in neuronal lysosomal lipid (NLL) accumulation, which opposed Aβ localization to lysosomes. Neuronal LAL loss both preceded (with aging) and promoted (targeted knockdown) Aβ pathology and cognitive deficits in AD mice. The addition of recombinant LAL ex vivo and neuronal LAL overexpression in vivo prevented amyloid increases and improved cognition. In WT mice, neuronal LAL declined with aging and correlated negatively with entorhinal Aβ. In healthy human brain, LAL also declined with age, suggesting this contributes to the age-related vulnerability for AD. In human LOAD LAL was further reduced, correlated negatively with Aβ1-42, and occurred with polymerase pausing at the LAL gene. Together, this finds that the loss of neuronal LAL promotes NLL accumulation to impede degradation of Aβ in neuronal lysosomes to drive AD amyloid pathology.
Collapse
Affiliation(s)
- Alexandra M Barnett
- University of North Carolina at Chapel Hill School of Medicine, Department of Pharmacology, Chapel Hill, NC
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC
| | - Lamar Dawkins
- University of North Carolina at Chapel Hill School of Medicine, Department of Pharmacology, Chapel Hill, NC
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC
| | - Jian Zou
- University of North Carolina at Chapel Hill School of Medicine, Department of Pharmacology, Chapel Hill, NC
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC
| | - Elizabeth McNair
- University of North Carolina at Chapel Hill School of Medicine, Department of Pharmacology, Chapel Hill, NC
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC
| | - Viktoriya D Nikolova
- University of North Carolina at Chapel Hill School of Medicine, Department of Psychiatry, Chapel Hill, NC
- University of North Carolina at Chapel Hill, Carolina Institute for Developmental Disabilities, Chapel Hill, NC
| | - Sheryl S Moy
- University of North Carolina at Chapel Hill School of Medicine, Department of Psychiatry, Chapel Hill, NC
- University of North Carolina at Chapel Hill, Carolina Institute for Developmental Disabilities, Chapel Hill, NC
| | - Greg T Sutherland
- New South Wales Brain Tissue Resource Centre and Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdon, Australia
| | - Julia Stevens
- New South Wales Brain Tissue Resource Centre and Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdon, Australia
| | - Meagan Colie
- University of North Carolina at Chapel Hill School of Medicine, Department of Pharmacology, Chapel Hill, NC
| | - Kemi Katemboh
- University of North Carolina at Chapel Hill School of Medicine, Department of Pharmacology, Chapel Hill, NC
| | - Hope Kellner
- University of North Carolina at Chapel Hill School of Medicine, Department of Pharmacology, Chapel Hill, NC
| | - Corina Damian
- University of North Carolina at Chapel Hill School of Medicine, Department of Pharmacology, Chapel Hill, NC
| | - Sagan DeCastro
- University of North Carolina at Chapel Hill School of Medicine, Department of Pharmacology, Chapel Hill, NC
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC
| | - Ryan P Vetreno
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC
- University of North Carolina at Chapel Hill School of Medicine, Department of Psychiatry, Chapel Hill, NC
| | - Leon G Coleman
- University of North Carolina at Chapel Hill School of Medicine, Department of Pharmacology, Chapel Hill, NC
- Bowles Center for Alcohol Studies, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC
| |
Collapse
|
68
|
Chamera K, Curzytek K, Kamińska K, Leśkiewicz M, Basta-Kaim A. Prenatal Immune Challenge Differentiates the Effect of Aripiprazole and Risperidone on CD200-CD200R and CX3CL1-CX3CR1 Dyads and Microglial Polarization: A Study in Organotypic Cortical Cultures. Life (Basel) 2024; 14:721. [PMID: 38929704 PMCID: PMC11205240 DOI: 10.3390/life14060721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/20/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
Microglia are the primary innate immune cells of the central nervous system and extensively contribute to brain homeostasis. Dysfunctional or excessive activity of microglia may be associated with several neuropsychiatric disorders, including schizophrenia. Therefore, we examined whether aripiprazole and risperidone could influence the expression of the Cd200-Cd200r and Cx3cl1-Cx3cr1 axes, which are crucial for the regulation of microglial activity and interactions of these cells with neurons. Additionally, we evaluated the impact of these drugs on microglial pro- and anti-inflammatory markers (Cd40, Il-1β, Il-6, Cebpb, Cd206, Arg1, Il-10 and Tgf-β) and cytokine release (IL-6, IL-10). The research was executed in organotypic cortical cultures (OCCs) prepared from the offspring of control rats (control OCCs) or those exposed to maternal immune activation (MIA OCCs), which allows for the exploration of schizophrenia-like disturbances in animals. All experiments were performed under basal conditions and after additional stimulation with lipopolysaccharide (LPS), following the "two-hit" hypothesis of schizophrenia. We found that MIA diminished the mRNA level of Cd200r and affected the OCCs' response to additional LPS exposure in terms of this parameter. LPS downregulated the Cx3cr1 expression and profoundly changed the mRNA levels of pro- and anti-inflammatory microglial markers in both types of OCCs. Risperidone increased Cd200 expression in MIA OCCs, while aripiprazole treatment elevated the gene levels of the Cx3cl1-Cx3cr1 dyad in control OCCs. The antipsychotics limited the LPS-generated increase in the expression of proinflammatory factors (Il-1β and Il-6) and enhanced the mRNA levels of anti-inflammatory components (Cd206 and Tgf-β) of microglial polarization, mostly in the absence of the MIA procedure. Finally, we observed a more pronounced modulating impact of aripiprazole on the expression of pro- and anti-inflammatory cytokines when compared to risperidone in MIA OCCs. In conclusion, our data suggest that MIA might influence microglial activation and crosstalk of microglial cells with neurons, whereas aripiprazole and risperidone could beneficially affect these changes in OCCs.
Collapse
Affiliation(s)
| | | | | | | | - Agnieszka Basta-Kaim
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna St., 31-343 Kraków, Poland
| |
Collapse
|
69
|
Plug BC, Revers IM, Breur M, González GM, Timmerman JA, Meijns NRC, Hamberg D, Wagendorp J, Nutma E, Wolf NI, Luchicchi A, Mansvelder HD, van Til NP, van der Knaap MS, Bugiani M. Human post-mortem organotypic brain slice cultures: a tool to study pathomechanisms and test therapies. Acta Neuropathol Commun 2024; 12:83. [PMID: 38822428 PMCID: PMC11140981 DOI: 10.1186/s40478-024-01784-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 04/16/2024] [Indexed: 06/03/2024] Open
Abstract
Human brain experimental models recapitulating age- and disease-related characteristics are lacking. There is urgent need for human-specific tools that model the complex molecular and cellular interplay between different cell types to assess underlying disease mechanisms and test therapies. Here we present an adapted ex vivo organotypic slice culture method using human post-mortem brain tissue cultured at an air-liquid interface to also study brain white matter. We assessed whether these human post-mortem brain slices recapitulate the in vivo neuropathology and if they are suitable for pathophysiological, experimental and pre-clinical treatment development purposes, specifically regarding leukodystrophies. Human post-mortem brain tissue and cerebrospinal fluid were obtained from control, psychiatric and leukodystrophy donors. Slices were cultured up to six weeks, in culture medium with or without human cerebrospinal fluid. Human post-mortem organotypic brain slice cultures remained viable for at least six weeks ex vivo and maintained tissue structure and diversity of (neural) cell types. Supplementation with cerebrospinal fluid could improve slice recovery. Patient-derived organotypic slice cultures recapitulated and maintained known in vivo neuropathology. The cultures also showed physiologic multicellular responses to lysolecithin-induced demyelination ex vivo, indicating their suitability to study intrinsic repair mechanisms upon injury. The slice cultures were applicable for various experimental studies, as multi-electrode neuronal recordings. Finally, the cultures showed successful cell-type dependent transduction with gene therapy vectors. These human post-mortem organotypic brain slice cultures represent an adapted ex vivo model suitable for multifaceted studies of brain disease mechanisms, boosting translation from human ex vivo to in vivo. This model also allows for assessing potential treatment options, including gene therapy applications. Human post-mortem brain slice cultures are thus a valuable tool in preclinical research to study the pathomechanisms of a wide variety of brain diseases in living human tissue.
Collapse
Affiliation(s)
- Bonnie C Plug
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Ilma M Revers
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Marjolein Breur
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Gema Muñoz González
- Department of Anatomy and Neurosciences, MS Center Amsterdam, Amsterdam University Medical Centre, VU University, Amsterdam Neuroscience, De Boelelaan 1108, Amsterdam, 1081 HZ, The Netherlands
| | - Jaap A Timmerman
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam Neuroscience, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Niels R C Meijns
- Department of Anatomy and Neurosciences, MS Center Amsterdam, Amsterdam University Medical Centre, VU University, Amsterdam Neuroscience, De Boelelaan 1108, Amsterdam, 1081 HZ, The Netherlands
| | - Daniek Hamberg
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Jikke Wagendorp
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Erik Nutma
- Department of Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
| | - Nicole I Wolf
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Antonio Luchicchi
- Department of Anatomy and Neurosciences, MS Center Amsterdam, Amsterdam University Medical Centre, VU University, Amsterdam Neuroscience, De Boelelaan 1108, Amsterdam, 1081 HZ, The Netherlands
| | - Huibert D Mansvelder
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam Neuroscience, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Niek P van Til
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam Neuroscience, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Marjo S van der Knaap
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam Neuroscience, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Marianna Bugiani
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands.
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands.
- Department of Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands.
| |
Collapse
|
70
|
Liu B, Buonomano DV. Ex Vivo Cortical Circuits Learn to Predict and Spontaneously Replay Temporal Patterns. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.30.596702. [PMID: 38853859 PMCID: PMC11160783 DOI: 10.1101/2024.05.30.596702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
It has been proposed that prediction and timing are computational primitives of neocortical microcircuits, specifically, that neural mechanisms are in place to allow neocortical circuits to autonomously learn the temporal structure of external stimuli and generate internal predictions. To test this hypothesis, we trained cortical organotypic slices on two specific temporal patterns using dual-optical stimulation. After 24-hours of training, whole-cell recordings revealed network dynamics consistent with training-specific timed prediction. Unexpectedly, there was replay of the learned temporal structure during spontaneous activity. Furthermore, some neurons exhibited timed prediction errors. Mechanistically our results indicate that learning relied in part on asymmetric connectivity between distinct neuronal ensembles with temporally-ordered activation. These findings further suggest that local cortical microcircuits are intrinsically capable of learning temporal information and generating predictions, and that the learning rules underlying temporal learning and spontaneous replay can be intrinsic to local cortical microcircuits and not necessarily dependent on top-down interactions.
Collapse
|
71
|
Flores JC, Zito K. A synapse-specific refractory period for plasticity at individual dendritic spines. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.24.595787. [PMID: 38826343 PMCID: PMC11142223 DOI: 10.1101/2024.05.24.595787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
How newly formed memories are preserved while brain plasticity is ongoing has been a source of debate. One idea is that synapses which experienced recent plasticity become resistant to further plasticity, a type of metaplasticity often referred to as saturation. Here, we probe the local dendritic mechanisms that limit plasticity at recently potentiated synapses. We show that recently potentiated individual synapses exhibit a synapse-specific refractory period for further potentiation. We further found that the refractory period is associated with reduced postsynaptic CaMKII signaling; however, stronger synaptic activation only partially restored the ability for further plasticity. Importantly, the refractory period is released after one hour, a timing that coincides with the enrichment of several postsynaptic proteins to pre-plasticity levels. Notably, increasing the level of the postsynaptic scaffolding protein, PSD95, but not of PSD93, overcomes the refractory period. Our results support a model in which potentiation at a single synapse is sufficient to initiate a synapse-specific refractory period that persists until key postsynaptic proteins regain their steady-state synaptic levels.
Collapse
Affiliation(s)
- Juan C. Flores
- Center for Neuroscience, University of California, Davis, CA 95618
| | - Karen Zito
- Center for Neuroscience, University of California, Davis, CA 95618
| |
Collapse
|
72
|
Aydin N, Ouliass B, Ferland G, Hafizi S. Modification of Gas6 Protein in the Brain by a Functional Endogenous Tissue Vitamin K Cycle. Cells 2024; 13:873. [PMID: 38786095 PMCID: PMC11119062 DOI: 10.3390/cells13100873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/06/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
The TAM receptor ligand Gas6 is known for regulating inflammatory and immune pathways in various organs including the brain. Gas6 becomes fully functional through the post-translational modification of multiple glutamic acid residues into γ-carboxyglutamic in a vitamin K-dependent manner. However, the significance of this mechanism in the brain is not known. We report here the endogenous expression of multiple components of the vitamin K cycle within the mouse brain at various ages as well as in distinct brain glial cells. The brain expression of all genes was increased in the postnatal ages, mirroring their profiles in the liver. In microglia, the proinflammatory agent lipopolysaccharide caused the downregulation of all key vitamin K cycle genes. A secreted Gas6 protein was detected in the medium of both mouse cerebellar slices and brain glial cell cultures. Furthermore, the endogenous Gas6 γ-carboxylation level was abolished through incubation with the vitamin K antagonist warfarin and could be restored through co-incubation with vitamin K1. Finally, the γ-carboxylation level of the Gas6 protein within the brains of warfarin-treated rats was found to be significantly reduced ex vivo compared to the control brains. In conclusion, we demonstrated for the first time the existence of a functional vitamin K cycle within rodent brains, which regulates the functional modification of endogenous brain Gas6. These results indicate that vitamin K is an important nutrient for the brain. Furthermore, the measurement of vitamin K-dependent Gas6 functionality could be an indicator of homeostatic or disease mechanisms in the brain, such as in neurological disorders where Gas6/TAM signalling is impaired.
Collapse
Affiliation(s)
- Nadide Aydin
- School of Medicine, Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK
| | - Bouchra Ouliass
- Département de Nutrition, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Guylaine Ferland
- Département de Nutrition, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Sassan Hafizi
- School of Medicine, Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK
| |
Collapse
|
73
|
Stoppini L, Heuschkel MO, Loussert-Fonta C, Gomez Baisac L, Roux A. Versatile micro-electrode array to monitor human iPSC derived 3D neural tissues at air-liquid interface. Front Cell Neurosci 2024; 18:1389580. [PMID: 38784710 PMCID: PMC11112036 DOI: 10.3389/fncel.2024.1389580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 04/02/2024] [Indexed: 05/25/2024] Open
Abstract
Engineered 3D neural tissues made of neurons and glial cells derived from human induced pluripotent stem cells (hiPSC) are among the most promising tools in drug discovery and neurotoxicology. They represent a cheaper, faster, and more ethical alternative to in vivo animal testing that will likely close the gap between in vitro animal models and human clinical trials. Micro-Electrode Array (MEA) technology is known to provide an assessment of compound effects on neural 2D cell cultures and acute tissue preparations by real-time, non-invasive, and long-lasting electrophysiological monitoring of spontaneous and evoked neuronal activity. Nevertheless, the use of engineered 3D neural tissues in combination with MEA biochips still involves series of constraints, such as drastically limited diffusion of oxygen and nutrients within tissues mainly due to the lack of vascularization. Therefore, 3D neural tissues are extremely sensitive to experimental conditions and require an adequately designed interface that provides optimal tissue survival conditions. A well-suited technique to overcome this issue is the combination of the Air-Liquid Interface (ALI) tissue culture method with the MEA technology. We have developed a full 3D neural tissue culture process and a data acquisition system composed of high-end electronics and novel MEA biochips based on porous, flexible, thin-film membranes integrating recording electrodes, named as "Strip-MEA," to allow the maintenance of an ALI around the 3D neural tissues. The main motivation of the porous MEA biochips development was the possibility to monitor and to study the electrical activity of 3D neural tissues under different recording configurations, (i) the Strip-MEA can be placed below a tissue, (ii) or by taking advantage of the ALI, be directly placed on top of the tissue, or finally, (iii) it can be embedded into a larger neural tissue generated by the fusion of two (or more) tissues placed on both sides of the Strip-MEA allowing the recording from its inner part. This paper presents the recording and analyses of spontaneous activity from the three positioning configurations of the Strip-MEAs. Obtained results are discussed with the perspective of developing in vitro models of brain diseases and/or impairment of neural network functioning.
Collapse
Affiliation(s)
| | | | | | | | - Adrien Roux
- Tissue Engineering Laboratory, HEPIA HES-SO University of Applied Sciences and Arts Western Switzerland, Geneva, Switzerland
| |
Collapse
|
74
|
Jordan FD, Kutter M, Comby JM, Brozzi F, Kurtys E. Open and remotely accessible Neuroplatform for research in wetware computing. Front Artif Intell 2024; 7:1376042. [PMID: 38756757 PMCID: PMC11097343 DOI: 10.3389/frai.2024.1376042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/11/2024] [Indexed: 05/18/2024] Open
Abstract
Wetware computing and organoid intelligence is an emerging research field at the intersection of electrophysiology and artificial intelligence. The core concept involves using living neurons to perform computations, similar to how Artificial Neural Networks (ANNs) are used today. However, unlike ANNs, where updating digital tensors (weights) can instantly modify network responses, entirely new methods must be developed for neural networks using biological neurons. Discovering these methods is challenging and requires a system capable of conducting numerous experiments, ideally accessible to researchers worldwide. For this reason, we developed a hardware and software system that allows for electrophysiological experiments on an unmatched scale. The Neuroplatform enables researchers to run experiments on neural organoids with a lifetime of even more than 100 days. To do so, we streamlined the experimental process to quickly produce new organoids, monitor action potentials 24/7, and provide electrical stimulations. We also designed a microfluidic system that allows for fully automated medium flow and change, thus reducing the disruptions by physical interventions in the incubator and ensuring stable environmental conditions. Over the past three years, the Neuroplatform was utilized with over 1,000 brain organoids, enabling the collection of more than 18 terabytes of data. A dedicated Application Programming Interface (API) has been developed to conduct remote research directly via our Python library or using interactive compute such as Jupyter Notebooks. In addition to electrophysiological operations, our API also controls pumps, digital cameras and UV lights for molecule uncaging. This allows for the execution of complex 24/7 experiments, including closed-loop strategies and processing using the latest deep learning or reinforcement learning libraries. Furthermore, the infrastructure supports entirely remote use. Currently in 2024, the system is freely available for research purposes, and numerous research groups have begun using it for their experiments. This article outlines the system's architecture and provides specific examples of experiments and results.
Collapse
|
75
|
Wise DL, Greene SB, Escobedo-Lozoya Y, Van Hooser SD, Nelson SB. Progressive Circuit Hyperexcitability in Mouse Neocortical Slice Cultures with Increasing Duration of Activity Silencing. eNeuro 2024; 11:ENEURO.0362-23.2024. [PMID: 38653560 PMCID: PMC11079856 DOI: 10.1523/eneuro.0362-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 04/09/2024] [Accepted: 04/12/2024] [Indexed: 04/25/2024] Open
Abstract
Forebrain neurons deprived of activity become hyperactive when activity is restored. Rebound activity has been linked to spontaneous seizures in vivo following prolonged activity blockade. Here, we measured the time course of rebound activity and the contributing circuit mechanisms using calcium imaging, synaptic staining, and whole-cell patch clamp in organotypic slice cultures of mouse neocortex. Calcium imaging revealed hypersynchronous activity increasing in intensity with longer periods of deprivation. While activity partially recovered 3 d after slices were released from 5 d of deprivation, they were less able to recover after 10 d of deprivation. However, even after the longer period of deprivation, activity patterns eventually returned to baseline levels. The degree of deprivation-induced rebound was age-dependent, with the greatest effects occurring when silencing began in the second week. Pharmacological blockade of NMDA receptors indicated that hypersynchronous rebound activity did not require activation of Hebbian plasticity. In single-neuron recordings, input resistance roughly doubled with a concomitant increase in intrinsic excitability. Synaptic imaging of pre- and postsynaptic proteins revealed dramatic reductions in the number of presumptive synapses with a larger effect on inhibitory than excitatory synapses. Putative excitatory synapses colocalizing PSD-95 and Bassoon declined by 39 and 56% following 5 and 10 d of deprivation, but presumptive inhibitory synapses colocalizing gephyrin and VGAT declined by 55 and 73%, respectively. The results suggest that with prolonged deprivation, a progressive reduction in synapse number is accompanied by a shift in the balance between excitation and inhibition and increased cellular excitability.
Collapse
Affiliation(s)
- Derek L Wise
- Department of Biology, Brandeis University, Waltham, Massachusetts 02454
| | - Samuel B Greene
- Department of Biology, Brandeis University, Waltham, Massachusetts 02454
| | | | | | - Sacha B Nelson
- Department of Biology, Brandeis University, Waltham, Massachusetts 02454
| |
Collapse
|
76
|
Petersilie L, Heiduschka S, Nelson JS, Neu LA, Le S, Anand R, Kafitz KW, Prigione A, Rose CR. Cortical brain organoid slices (cBOS) for the study of human neural cells in minimal networks. iScience 2024; 27:109415. [PMID: 38523789 PMCID: PMC10957451 DOI: 10.1016/j.isci.2024.109415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 01/29/2024] [Accepted: 02/29/2024] [Indexed: 03/26/2024] Open
Abstract
Brain organoids derived from human pluripotent stem cells are a promising tool for studying human neurodevelopment and related disorders. Here, we generated long-term cultures of cortical brain organoid slices (cBOS) grown at the air-liquid interphase from regionalized cortical organoids. We show that cBOS host mature neurons and astrocytes organized in complex architecture. Whole-cell patch-clamp demonstrated subthreshold synaptic inputs and action potential firing of neurons. Spontaneous intracellular calcium signals turned into synchronous large-scale oscillations upon combined disinhibition of NMDA receptors and blocking of GABAA receptors. Brief metabolic inhibition to mimic transient energy restriction in the ischemic brain induced reversible intracellular calcium loading of cBOS. Moreover, metabolic inhibition induced a reversible decline in neuronal ATP as revealed by ATeam1.03YEMK. Overall, cBOS provide a powerful platform to assess morphological and functional aspects of human neural cells in intact minimal networks and to address the pathways that drive cellular damage during brain ischemia.
Collapse
Affiliation(s)
- Laura Petersilie
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Sonja Heiduschka
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children’s Hospital and Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Joel S.E. Nelson
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Louis A. Neu
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Stephanie Le
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children’s Hospital and Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Ruchika Anand
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Duesseldorf, Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Karl W. Kafitz
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Alessandro Prigione
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children’s Hospital and Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Christine R. Rose
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| |
Collapse
|
77
|
Zhao K, Braun M, Meyer L, Otte K, Raifer H, Helmprobst F, Möschl V, Pagenstecher A, Urban H, Ronellenfitsch MW, Steinbach JP, Pesek J, Watzer B, Nockher WA, Taudte RV, Neubauer A, Nimsky C, Bartsch JW, Rusch T. A Novel Approach for Glioblastoma Treatment by Combining Apoptosis Inducers (TMZ, MTX, and Cytarabine) with E.V.A. (Eltanexor, Venetoclax, and A1210477) Inhibiting XPO1, Bcl-2, and Mcl-1. Cells 2024; 13:632. [PMID: 38607071 PMCID: PMC11011525 DOI: 10.3390/cells13070632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024] Open
Abstract
Adjuvant treatment for Glioblastoma Grade 4 with Temozolomide (TMZ) inevitably fails due to therapeutic resistance, necessitating new approaches. Apoptosis induction in GB cells is inefficient, due to an excess of anti-apoptotic XPO1/Bcl-2-family proteins. We assessed TMZ, Methotrexate (MTX), and Cytarabine (Ara-C) (apoptosis inducers) combined with XPO1/Bcl-2/Mcl-1-inhibitors (apoptosis rescue) in GB cell lines and primary GB stem-like cells (GSCs). Using CellTiter-Glo® and Caspase-3 activity assays, we generated dose-response curves and analyzed the gene and protein regulation of anti-apoptotic proteins via PCR and Western blots. Optimal drug combinations were examined for their impact on the cell cycle and apoptosis induction via FACS analysis, paralleled by the assessment of potential toxicity in healthy mouse brain slices. Ara-C and MTX proved to be 150- to 10,000-fold more potent in inducing apoptosis than TMZ. In response to inhibitors Eltanexor (XPO1; E), Venetoclax (Bcl-2; V), and A1210477 (Mcl-1; A), genes encoding for the corresponding proteins were upregulated in a compensatory manner. TMZ, MTX, and Ara-C combined with E, V, and A evidenced highly lethal effects when combined. As no significant cell death induction in mouse brain slices was observed, we conclude that this drug combination is effective in vitro and expected to have low side effects in vivo.
Collapse
Affiliation(s)
- Kai Zhao
- Department of Neurosurgery, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
- Department of Hematology, Oncology & Immunology, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
| | - Madita Braun
- Department of Neurosurgery, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
- Department of Hematology, Oncology & Immunology, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
| | - Leonie Meyer
- Department of Neurosurgery, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
- Department of Hematology, Oncology & Immunology, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
| | - Katharina Otte
- Department of Neurosurgery, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
- Department of Hematology, Oncology & Immunology, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
| | - Hartmann Raifer
- FACS Core Facility, Philipps University Marburg, Hans-Meerwein-Straße 3, 35043 Marburg, Germany
| | - Frederik Helmprobst
- Department of Neuropathology, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
| | - Vincent Möschl
- Department of Neuropathology, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
| | - Axel Pagenstecher
- Department of Neuropathology, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
- University Cancer Center (UCT) Frankfurt—Marburg, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Hans Urban
- University Cancer Center (UCT) Frankfurt—Marburg, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- Dr. Senckenberg Institute of Neurooncology, Goethe-University of Frankfurt, Schleusenweg 2-16, 60528 Frankfurt am Main, Germany
| | - Michael W. Ronellenfitsch
- University Cancer Center (UCT) Frankfurt—Marburg, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- Dr. Senckenberg Institute of Neurooncology, Goethe-University of Frankfurt, Schleusenweg 2-16, 60528 Frankfurt am Main, Germany
| | - Joachim P. Steinbach
- University Cancer Center (UCT) Frankfurt—Marburg, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- Dr. Senckenberg Institute of Neurooncology, Goethe-University of Frankfurt, Schleusenweg 2-16, 60528 Frankfurt am Main, Germany
| | - Jelena Pesek
- Medical Mass Spectrometry Core Facility, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
| | - Bernhard Watzer
- Medical Mass Spectrometry Core Facility, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
| | - Wolfgang A. Nockher
- Medical Mass Spectrometry Core Facility, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
| | - R. Verena Taudte
- Medical Mass Spectrometry Core Facility, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
| | - Andreas Neubauer
- Department of Hematology, Oncology & Immunology, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
- University Cancer Center (UCT) Frankfurt—Marburg, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Christopher Nimsky
- Department of Neurosurgery, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
- University Cancer Center (UCT) Frankfurt—Marburg, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Jörg W. Bartsch
- Department of Neurosurgery, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
- University Cancer Center (UCT) Frankfurt—Marburg, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Tillmann Rusch
- Department of Hematology, Oncology & Immunology, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
- University Cancer Center (UCT) Frankfurt—Marburg, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| |
Collapse
|
78
|
Rao S, Sadybekov A, DeWitt DC, Lipka J, Katritch V, Herring BE. Detection of autism spectrum disorder-related pathogenic trio variants by a novel structure-based approach. Mol Autism 2024; 15:12. [PMID: 38566250 PMCID: PMC10988830 DOI: 10.1186/s13229-024-00590-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/16/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Glutamatergic synapse dysfunction is believed to underlie the development of Autism Spectrum Disorder (ASD) and Intellectual Disability (ID) in many individuals. However, identification of genetic markers that contribute to synaptic dysfunction in these individuals is notoriously difficult. Based on genomic analysis, structural modeling, and functional data, we recently established the involvement of the TRIO-RAC1 pathway in ASD and ID. Furthermore, we identified a pathological de novo missense mutation hotspot in TRIO's GEF1 domain. ASD/ID-related missense mutations within this domain compromise glutamatergic synapse function and likely contribute to the development of ASD/ID. The number of ASD/ID cases with mutations identified within TRIO's GEF1 domain is increasing. However, tools for accurately predicting whether such mutations are detrimental to protein function are lacking. METHODS Here we deployed advanced protein structural modeling techniques to predict potential de novo pathogenic and benign mutations within TRIO's GEF1 domain. Mutant TRIO-9 constructs were generated and expressed in CA1 pyramidal neurons of organotypic cultured hippocampal slices. AMPA receptor-mediated postsynaptic currents were examined in these neurons using dual whole-cell patch clamp electrophysiology. We also validated these findings using orthogonal co-immunoprecipitation and fluorescence lifetime imaging (FLIM-FRET) experiments to assay TRIO mutant overexpression effects on TRIO-RAC1 binding and on RAC1 activity in HEK293/T cells. RESULTS Missense mutations in TRIO's GEF1 domain that were predicted to disrupt TRIO-RAC1 binding or stability were tested experimentally and found to greatly impair TRIO-9's influence on glutamatergic synapse function. In contrast, missense mutations in TRIO's GEF1 domain that were predicted to have minimal effect on TRIO-RAC1 binding or stability did not impair TRIO-9's influence on glutamatergic synapse function in our experimental assays. In orthogonal assays, we find most of the mutations predicted to disrupt binding display loss of function but mutants predicted to disrupt stability do not reflect our results from neuronal electrophysiological data. LIMITATIONS We present a method to predict missense mutations in TRIO's GEF1 domain that may compromise TRIO function and test for effects in a limited number of assays. Possible limitations arising from the model systems employed here can be addressed in future studies. Our method does not provide evidence for whether these mutations confer ASD/ID risk or the likelihood that such mutations will result in the development of ASD/ID. CONCLUSIONS Here we show that a combination of structure-based computational predictions and experimental validation can be employed to reliably predict whether missense mutations in the human TRIO gene impede TRIO protein function and compromise TRIO's role in glutamatergic synapse regulation. With the growing accessibility of genome sequencing, the use of such tools in the accurate identification of pathological mutations will be instrumental in diagnostics of ASD/ID.
Collapse
Affiliation(s)
- Sadhna Rao
- Department of Biological Sciences, Neurobiology Section, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, 90089, USA.
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, 90089, USA.
| | - Anastasiia Sadybekov
- Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, 90089, USA
- Department of Chemistry, University of Southern California, Los Angeles, CA, 90089, USA
| | - David C DeWitt
- Department of Pathology, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Joanna Lipka
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA, 94080, USA
| | - Vsevolod Katritch
- Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, 90089, USA.
- Department of Chemistry, University of Southern California, Los Angeles, CA, 90089, USA.
| | - Bruce E Herring
- Department of Biological Sciences, Neurobiology Section, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, 90089, USA.
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, 90089, USA.
| |
Collapse
|
79
|
Heir R, Abbasi Z, Komal P, Altimimi HF, Franquin M, Moschou D, Chambon J, Stellwagen D. Astrocytes Are the Source of TNF Mediating Homeostatic Synaptic Plasticity. J Neurosci 2024; 44:e2278222024. [PMID: 38395613 PMCID: PMC10993029 DOI: 10.1523/jneurosci.2278-22.2024] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Tumor necrosis factor α (TNF) mediates homeostatic synaptic plasticity (HSP) in response to chronic activity blockade, and prior work has established that it is released from glia. Here we demonstrate that astrocytes are the necessary source of TNF during HSP. Hippocampal cultures from rats of both sexes depleted of microglia still will increase TNF levels following activity deprivation and still express TTX-driven HSP. Slice cultures from mice of either sex with a conditional deletion of TNF from microglia also express HSP, but critically, slice cultures with a conditional deletion of TNF from astrocytes do not. In astrocytes, glutamate signaling is sufficient to reduce NFκB signaling and TNF mRNA levels. Further, chronic TTX treatment increases TNF in an NFκB-dependent manner, although NFκB signaling is dispensable for the neuronal response to TTX-driven HSP. Thus, astrocytes can sense neuronal activity through glutamate spillover and increase TNF production when activity falls, to drive HSP through the production of TNF.
Collapse
Affiliation(s)
- Renu Heir
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, Quebec H3G 1A4, Canada
| | - Zahra Abbasi
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, Quebec H3G 1A4, Canada
| | - Pragya Komal
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, Quebec H3G 1A4, Canada
| | - Haider F Altimimi
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, Quebec H3G 1A4, Canada
| | - Marie Franquin
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, Quebec H3G 1A4, Canada
| | - Dionysia Moschou
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, Quebec H3G 1A4, Canada
| | - Julien Chambon
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, Quebec H3G 1A4, Canada
| | - David Stellwagen
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, Quebec H3G 1A4, Canada
| |
Collapse
|
80
|
Choquet D, Petrel M, Fernández-Monreal M. Targeting of membrane proteins with fluoronanogold probes for high-resolution correlative microscopy. Methods Cell Biol 2024; 187:57-72. [PMID: 38705630 DOI: 10.1016/bs.mcb.2024.02.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
Correlative light and electron microscopy (CLEM) can provide valuable information about a biological sample by giving information on the specific localization of a molecule of interest within an ultrastructural context. In this work, we describe a simple CLEM method to obtain high-resolution images of neurotransmitter receptor distribution in synapses by electron microscopy (EM). We use hippocampal organotypic slices from a previously reported mouse model expressing a modified AMPA receptor (AMPAR) subunit that binds biotin at the surface (Getz et al., 2022). This tag can be recognized by StreptAvidin-Fluoronanogold™ conjugates (SA-FNG), which reach receptors at synapses (synaptic cleft is 50-100nm thick). By using pre-embedding labeling, we found that SA-FNG reliably bind synaptic receptors and penetrate around 10-15μm in depth in live tissue. However, the silver enhancement was only reaching the surface of the slices. We show that permeabilization with triton is highly effective at increasing the in depth-gold amplification and that the membrane integrity is well preserved. Finally, we also apply high-resolution electron tomography, thus providing important information about the 3D organization of surface AMPA receptors in synapses at the nanoscale.
Collapse
Affiliation(s)
- Daniel Choquet
- Université de Bordeaux, CNRS, INSERM, Bordeaux Imaging Center (BIC), Bordeaux, France; Université de Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience (IINS), Bordeaux, France
| | - Melina Petrel
- Université de Bordeaux, CNRS, INSERM, Bordeaux Imaging Center (BIC), Bordeaux, France
| | | |
Collapse
|
81
|
Hartmann J, Bajaj T, Otten J, Klengel C, Ebert T, Gellner AK, Junglas E, Hafner K, Anderzhanova EA, Tang F, Missig G, Rexrode L, Trussell DT, Li KX, Pöhlmann ML, Mackert S, Geiger TM, Heinz DE, Lardenoije R, Dedic N, McCullough KM, Próchnicki T, Rhomberg T, Martinelli S, Payton A, Robinson AC, Stein V, Latz E, Carlezon WA, Hausch F, Schmidt MV, Murgatroyd C, Berretta S, Klengel T, Pantazopoulos H, Ressler KJ, Gassen NC. SKA2 regulated hyperactive secretory autophagy drives neuroinflammation-induced neurodegeneration. Nat Commun 2024; 15:2635. [PMID: 38528004 PMCID: PMC10963788 DOI: 10.1038/s41467-024-46953-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/15/2024] [Indexed: 03/27/2024] Open
Abstract
High levels of proinflammatory cytokines induce neurotoxicity and catalyze inflammation-driven neurodegeneration, but the specific release mechanisms from microglia remain elusive. Here we show that secretory autophagy (SA), a non-lytic modality of autophagy for secretion of vesicular cargo, regulates neuroinflammation-mediated neurodegeneration via SKA2 and FKBP5 signaling. SKA2 inhibits SA-dependent IL-1β release by counteracting FKBP5 function. Hippocampal Ska2 knockdown in male mice hyperactivates SA resulting in neuroinflammation, subsequent neurodegeneration and complete hippocampal atrophy within six weeks. The hyperactivation of SA increases IL-1β release, contributing to an inflammatory feed-forward vicious cycle including NLRP3-inflammasome activation and Gasdermin D-mediated neurotoxicity, which ultimately drives neurodegeneration. Results from protein expression and co-immunoprecipitation analyses of male and female postmortem human brains demonstrate that SA is hyperactivated in Alzheimer's disease. Overall, our findings suggest that SKA2-regulated, hyperactive SA facilitates neuroinflammation and is linked to Alzheimer's disease, providing mechanistic insight into the biology of neuroinflammation.
Collapse
Affiliation(s)
- Jakob Hartmann
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA.
| | - Thomas Bajaj
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, 53127, Bonn, Germany
| | - Joy Otten
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, 37075, Göttingen, Germany
| | - Claudia Klengel
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Tim Ebert
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, 53127, Bonn, Germany
| | - Anne-Kathrin Gellner
- Department of Psychiatry and Psychotherapy, University of Bonn, 53127, Bonn, Germany
| | - Ellen Junglas
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, 53127, Bonn, Germany
| | - Kathrin Hafner
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Elmira A Anderzhanova
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, 53127, Bonn, Germany
- Research Group Neuronal Plasticity, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Fiona Tang
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Galen Missig
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Lindsay Rexrode
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Daniel T Trussell
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Katelyn X Li
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Max L Pöhlmann
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Sarah Mackert
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, 53127, Bonn, Germany
| | - Thomas M Geiger
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, 64287, Darmstadt, Germany
| | - Daniel E Heinz
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, 53127, Bonn, Germany
- Research Group Neuronal Plasticity, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Roy Lardenoije
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, 37075, Göttingen, Germany
| | - Nina Dedic
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Kenneth M McCullough
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Tomasz Próchnicki
- Institute of Innate Immunity, University Hospital Bonn, 53127, Bonn, Germany
| | - Thomas Rhomberg
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Silvia Martinelli
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Antony Payton
- Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, M13 9PL, UK
| | - Andrew C Robinson
- Division of Neuroscience, Faculty of Biology, Medicine and Health, School of Biological Sciences, The University of Manchester, Salford Royal Hospital, Salford, M6 8HD, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre (MAHSC), Manchester, UK
| | - Valentin Stein
- Institute of Physiology II, University of Bonn, 53127, Bonn, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital Bonn, 53127, Bonn, Germany
- Deutsches Rheuma Forschungszentrum Berlin (DRFZ), 10117, Berlin, Germany
| | - William A Carlezon
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Felix Hausch
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, 64287, Darmstadt, Germany
| | - Mathias V Schmidt
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Chris Murgatroyd
- Department of Life Sciences, Manchester Metropolitan University, Manchester, M15 6BH, UK
| | - Sabina Berretta
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
| | - Torsten Klengel
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, 37075, Göttingen, Germany
| | - Harry Pantazopoulos
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Kerry J Ressler
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, 02478, USA.
| | - Nils C Gassen
- Research Group Neurohomeostasis, Department of Psychiatry and Psychotherapy, University of Bonn, 53127, Bonn, Germany.
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804, Munich, Germany.
| |
Collapse
|
82
|
Anjum R, Clarke VRJ, Nagasawa Y, Murakoshi H, Paradis S. Rem2 interacts with CaMKII at synapses and restricts long-term potentiation in hippocampus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.11.584540. [PMID: 38558974 PMCID: PMC10979978 DOI: 10.1101/2024.03.11.584540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Synaptic plasticity, the process whereby neuronal connections are either strengthened or weakened in response to stereotyped forms of stimulation, is widely believed to represent the molecular mechanism that underlies learning and memory. The holoenzyme CaMKII plays a well-established and critical role in the induction of a variety of forms of synaptic plasticity such as long-term potentiation (LTP), long-term depression (LTD) and depotentiation. Previously, we identified the GTPase Rem2 as a potent, endogenous inhibitor of CaMKII. Here, we report that knock out of Rem2 enhances LTP at the Schaffer collateral to CA1 synapse in hippocampus, consistent with an inhibitory action of Rem2 on CaMKII in vivo. Further, re-expression of WT Rem2 rescues the enhanced LTP observed in slices obtained from Rem2 conditional knock out (cKO) mice, while expression of a mutant Rem2 construct that is unable to inhibit CaMKII in vitro fails to rescue increased LTP. In addition, we demonstrate that CaMKII and Rem2 interact in dendritic spines using a 2pFLIM-FRET approach. Taken together, our data lead us to propose that Rem2 serves as a brake on runaway synaptic potentiation via inhibition of CaMKII activity. Further, the enhanced LTP phenotype we observe in Rem2 cKO slices reveals a previously unknown role for Rem2 in the negative regulation of CaMKII function.
Collapse
Affiliation(s)
- Rabia Anjum
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, Massachusetts 02454, United States of America
| | - Vernon R J Clarke
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Yutaro Nagasawa
- Department of Physiological Sciences, The Graduate University for Advanced Studies; Hayama, Kanagawa 240-0193, Japan
- Supportive Center for Brain Research, National Institute for Physiological Sciences; Okazaki, Aichi 444-8585, Japan
| | - Hideji Murakoshi
- Department of Physiological Sciences, The Graduate University for Advanced Studies; Hayama, Kanagawa 240-0193, Japan
- Supportive Center for Brain Research, National Institute for Physiological Sciences; Okazaki, Aichi 444-8585, Japan
| | - Suzanne Paradis
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, Massachusetts 02454, United States of America
| |
Collapse
|
83
|
Adel SS, Pranske ZJ, Kowalski TF, Kanzler N, Ray R, Carmona C, Paradis S. Plexin-B1 and Plexin-B2 play non-redundant roles in GABAergic synapse formation. Mol Cell Neurosci 2024; 128:103920. [PMID: 38331011 PMCID: PMC11046529 DOI: 10.1016/j.mcn.2024.103920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/24/2024] [Accepted: 01/28/2024] [Indexed: 02/10/2024] Open
Abstract
Synapse formation in the mammalian brain is a complex and dynamic process requiring coordinated function of dozens of molecular families such as cell adhesion molecules (CAMs) and ligand-receptor pairs (Ephs/Ephrins, Neuroligins/Neurexins, Semaphorins/Plexins). Due to the large number of molecular players and possible functional redundancies within gene families, it is challenging to determine the precise synaptogenic roles of individual molecules, which is key to understanding the consequences of mutations in these genes for brain function. Furthermore, few molecules are known to exclusively regulate either GABAergic or glutamatergic synapses, and cell and molecular mechanisms underlying GABAergic synapse formation in particular are not thoroughly understood. We previously demonstrated that Semaphorin-4D (Sema4D) regulates GABAergic synapse development in the mammalian hippocampus while having no effect on glutamatergic synapse development, and this effect occurs through binding to its high affinity receptor, Plexin-B1. In addition, we demonstrated that RNAi-mediated Plexin-B2 knock-down decreases GABAergic synapse density suggesting that both receptors function in this process. Here, we perform a structure-function study of the Plexin-B1 and Plexin-B2 receptors to identify the protein domains in each receptor which are required for its synaptogenic function. Further, we examine whether Plexin-B2 is required in the presynaptic neuron, the postsynaptic neuron, or both to regulate GABAergic synapse formation. Our data reveal that Plexin-B1 and Plexin-B2 function non-redundantly to regulate GABAergic synapse formation and suggest that the transmembrane domain may underlie functional distinctions. We also provide evidence that Plexin-B2 expression in presynaptic GABAergic interneurons, as well as postsynaptic pyramidal cells, regulates GABAergic synapse formation in hippocampus. These findings lay the groundwork for future investigations into the precise signaling pathways required for synapse formation downstream of Plexin-B receptor signaling.
Collapse
Affiliation(s)
- Susannah S Adel
- Department of Biology, Brandeis University, Waltham, MA 02454, United States of America
| | - Zachary J Pranske
- Department of Biology, Brandeis University, Waltham, MA 02454, United States of America
| | - Tess F Kowalski
- Department of Biology, Brandeis University, Waltham, MA 02454, United States of America
| | - Nicole Kanzler
- Department of Biology, Brandeis University, Waltham, MA 02454, United States of America
| | - Roshni Ray
- Department of Biology, Brandeis University, Waltham, MA 02454, United States of America
| | - Catherine Carmona
- Department of Biology, Brandeis University, Waltham, MA 02454, United States of America
| | - Suzanne Paradis
- Department of Biology, Brandeis University, Waltham, MA 02454, United States of America.
| |
Collapse
|
84
|
Kiy Z, Chaud J, Xu L, Brandhorst E, Kamali T, Vargas C, Keller S, Hong H, Specht A, Cambridge S. Towards a Light-mediated Gene Therapy for the Eye using Caged Ethinylestradiol and the Inducible Cre/lox System. Angew Chem Int Ed Engl 2024; 63:e202317675. [PMID: 38127455 DOI: 10.1002/anie.202317675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023]
Abstract
Increasingly, retinal pathologies are being treated with virus-mediated gene therapies. To be able to target viral transgene expression specifically to the pathological regions of the retina with light, we established an in vivo photoactivated gene expression paradigm for retinal tissue. Based on the inducible Cre/lox system, we discovered that ethinylestradiol is a suitable alternative to Tamoxifen as ethinylestradiol is more amenable to modification with photosensitive protecting compounds, i.e., "caging." Identification of ethinylestradiol as a ligand for the mutated human estradiol receptor was supported by in silico binding studies showing the reduced binding of caged ethinylestradiol. Caged ethinylestradiol was injected into the eyes of double transgenic GFAP-CreERT2 mice with a Cre-dependent tdTomato reporter transgene followed by irradiation with light of 450 nm. Photoactivation significantly increased retinal tdTomato expression compared to controls. We thus demonstrated a first step towards the development of a targeted, light-mediated gene therapy for the eyes.
Collapse
Affiliation(s)
- Zoe Kiy
- Heidelberg University, 69120, Heidelberg, Germany
| | - Juliane Chaud
- Laboratoire de Conception et Application de Molécules Bioactives, Equipe de Chimie et Neurobiologie Moléculaire, Université de Strasbourg, CNRS, CAMB UMR 7199, 67000, Strasbourg, France
| | - Liang Xu
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Eric Brandhorst
- Sektion Endokrinologie, Medizinische Fakultät Mannheim, 68167, Mannheim, Germany
| | - Tschackad Kamali
- Heidelberg Engineering GmbH, Max-Jarecki-Straße 8, 69115, Heidelberg, Germany
| | - Carolyn Vargas
- Biophysics, Institute of Molecular Biosciences (IMB), NAWI Graz, University of Graz, Humboldtstr. 50/III, 8010, Graz, Austria
- BioTechMed-Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
| | - Sandro Keller
- Biophysics, Institute of Molecular Biosciences (IMB), NAWI Graz, University of Graz, Humboldtstr. 50/III, 8010, Graz, Austria
- BioTechMed-Graz, Graz, Austria
- Field of Excellence BioHealth, University of Graz, Graz, Austria
| | - Huixiao Hong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Alexandre Specht
- Laboratoire de Conception et Application de Molécules Bioactives, Equipe de Chimie et Neurobiologie Moléculaire, Université de Strasbourg, CNRS, CAMB UMR 7199, 67000, Strasbourg, France
| | - Sidney Cambridge
- Heidelberg University, 69120, Heidelberg, Germany
- Institute for Anatomy II, Dr. Senckenberg Anatomy, Goethe-University Frankfurt am Main, 60590, Frankfurt am Main, Germany
| |
Collapse
|
85
|
Ogelman R, Gomez Wulschner LE, Hoelscher VM, Hwang IW, Chang VN, Oh WC. Serotonin modulates excitatory synapse maturation in the developing prefrontal cortex. Nat Commun 2024; 15:1368. [PMID: 38365905 PMCID: PMC10873381 DOI: 10.1038/s41467-024-45734-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 02/02/2024] [Indexed: 02/18/2024] Open
Abstract
Serotonin (5-HT) imbalances in the developing prefrontal cortex (PFC) are linked to long-term behavioral deficits. However, the synaptic mechanisms underlying 5-HT-mediated PFC development are unknown. We found that chemogenetic suppression and enhancement of 5-HT release in the PFC during the first two postnatal weeks decreased and increased the density and strength of excitatory spine synapses, respectively, on prefrontal layer 2/3 pyramidal neurons in mice. 5-HT release on single spines induced structural and functional long-term potentiation (LTP), requiring both 5-HT2A and 5-HT7 receptor signals, in a glutamatergic activity-independent manner. Notably, LTP-inducing 5-HT stimuli increased the long-term survival of newly formed spines ( ≥ 6 h) via 5-HT7 Gαs activation. Chronic treatment of mice with fluoxetine, a selective serotonin-reuptake inhibitor, during the first two weeks, but not the third week of postnatal development, increased the density and strength of excitatory synapses. The effect of fluoxetine on PFC synaptic alterations in vivo was abolished by 5-HT2A and 5-HT7 receptor antagonists. Our data describe a molecular basis of 5-HT-dependent excitatory synaptic plasticity at the level of single spines in the PFC during early postnatal development.
Collapse
Affiliation(s)
- Roberto Ogelman
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Luis E Gomez Wulschner
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Victoria M Hoelscher
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - In-Wook Hwang
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Victoria N Chang
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Won Chan Oh
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA.
| |
Collapse
|
86
|
Inavalli VVGK, Puente Muñoz V, Draffin JE, Tønnesen J. Fluorescence microscopy shadow imaging for neuroscience. Front Cell Neurosci 2024; 18:1330100. [PMID: 38425431 PMCID: PMC10902105 DOI: 10.3389/fncel.2024.1330100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 02/01/2024] [Indexed: 03/02/2024] Open
Abstract
Fluorescence microscopy remains one of the single most widely applied experimental approaches in neuroscience and beyond and is continuously evolving to make it easier and more versatile. The success of the approach is based on synergistic developments in imaging technologies and fluorophore labeling strategies that have allowed it to greatly diversify and be used across preparations for addressing structure as well as function. Yet, while targeted labeling strategies are a key strength of fluorescence microscopy, they reciprocally impose general limitations on the possible types of experiments and analyses. One recent development that overcomes some of these limitations is fluorescence microscopy shadow imaging, where membrane-bound cellular structures remain unlabeled while the surrounding extracellular space is made to fluoresce to provide a negative contrast shadow image. When based on super-resolution STED microscopy, the technique in effect provides a positive image of the extracellular space geometry and entire neuropil in the field of view. Other noteworthy advantages include the near elimination of the adverse effects of photobleaching and toxicity in live imaging, exhaustive and homogeneous labeling across the preparation, and the ability to apply and adjust the label intensity on the fly. Shadow imaging is gaining popularity and has been applied on its own or combined with conventional positive labeling to visualize cells and synaptic proteins in their parenchymal context. Here, we highlight the inherent limitations of fluorescence microscopy and conventional labeling and contrast these against the pros and cons of recent shadow imaging approaches. Our aim is to describe the brief history and current trajectory of the shadow imaging technique in the neuroscience field, and to draw attention to its ease of application and versatility.
Collapse
Affiliation(s)
| | - Virginia Puente Muñoz
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
- Neuronal Excitability Lab, Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Jonathan E. Draffin
- Neuronal Excitability Lab, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Aligning Science Across Parkinson’s (ASAP), Collaborative Research Network, Chevy Chase, MD, United States
| | - Jan Tønnesen
- Department of Neurosciences, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
- Neuronal Excitability Lab, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Aligning Science Across Parkinson’s (ASAP), Collaborative Research Network, Chevy Chase, MD, United States
- Instituto Biofisika (CSIC/UPV), Leioa, Spain
| |
Collapse
|
87
|
Fernholz MHP, Guggiana Nilo DA, Bonhoeffer T, Kist AM. DeepD3, an open framework for automated quantification of dendritic spines. PLoS Comput Biol 2024; 20:e1011774. [PMID: 38422112 PMCID: PMC10903918 DOI: 10.1371/journal.pcbi.1011774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 12/20/2023] [Indexed: 03/02/2024] Open
Abstract
Dendritic spines are the seat of most excitatory synapses in the brain, and a cellular structure considered central to learning, memory, and activity-dependent plasticity. The quantification of dendritic spines from light microscopy data is usually performed by humans in a painstaking and error-prone process. We found that human-to-human variability is substantial (inter-rater reliability 82.2±6.4%), raising concerns about the reproducibility of experiments and the validity of using human-annotated 'ground truth' as an evaluation method for computational approaches of spine identification. To address this, we present DeepD3, an open deep learning-based framework to robustly quantify dendritic spines in microscopy data in a fully automated fashion. DeepD3's neural networks have been trained on data from different sources and experimental conditions, annotated and segmented by multiple experts and they offer precise quantification of dendrites and dendritic spines. Importantly, these networks were validated in a number of datasets on varying acquisition modalities, species, anatomical locations and fluorescent indicators. The entire DeepD3 open framework, including the fully segmented training data, a benchmark that multiple experts have annotated, and the DeepD3 model zoo is fully available, addressing the lack of openly available datasets of dendritic spines while offering a ready-to-use, flexible, transparent, and reproducible spine quantification method.
Collapse
Affiliation(s)
| | | | - Tobias Bonhoeffer
- Max-Planck-Institute for Biological Intelligence, Martinsried, Bavaria, Germany
| | - Andreas M. Kist
- Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Bavaria, Germany
| |
Collapse
|
88
|
Larson KC, Martens LH, Marconi M, Dejesus C, Bruhn S, Miller TA, Tate B, Levenson JM. Preclinical translational platform of neuroinflammatory disease biology relevant to neurodegenerative disease. J Neuroinflammation 2024; 21:37. [PMID: 38297405 PMCID: PMC10832185 DOI: 10.1186/s12974-024-03029-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 01/23/2024] [Indexed: 02/02/2024] Open
Abstract
Neuroinflammation is a key driver of neurodegenerative disease, however the tools available to model this disease biology at the systems level are lacking. We describe a translational drug discovery platform based on organotypic culture of murine cortical brain slices that recapitulate disease-relevant neuroinflammatory biology. After an acute injury response, the brain slices assume a chronic neuroinflammatory state marked by transcriptomic profiles indicative of activation of microglia and astrocytes and loss of neuronal function. Microglia are necessary for manifestation of this neuroinflammation, as depletion of microglia prior to isolation of the brain slices prevents both activation of astrocytes and robust loss of synaptic function genes. The transcriptomic pattern of neuroinflammation in the mouse platform is present in published datasets derived from patients with amyotrophic lateral sclerosis, Huntington's disease, and frontotemporal dementia. Pharmacological utility of the platform was validated by demonstrating reversal of microglial activation and the overall transcriptomic signature with transforming growth factor-β. Additional anti-inflammatory targets were screened and inhibitors of glucocorticoid receptors, COX-2, dihydrofolate reductase, and NLRP3 inflammasome all failed to reverse the neuroinflammatory signature. Bioinformatics analysis of the neuroinflammatory signature identified protein tyrosine phosphatase non-receptor type 11 (PTPN11/SHP2) as a potential target. Three structurally distinct inhibitors of PTPN11 (RMC-4550, TN0155, IACS-13909) reversed the neuroinflammatory disease signature. Collectively, these results highlight the utility of this novel neuroinflammatory platform for facilitating identification and validation of targets for neuroinflammatory neurodegenerative disease drug discovery.
Collapse
Affiliation(s)
- Kelley C Larson
- Vigil Neuroscience, Watertown, USA
- Tiaki Therapeutics, Inc., c/o Dementia Discovery Fund, 201 Washington Street, 39th Floor, Boston, MA, 02108, USA
| | - Lauren H Martens
- , Neumora Therapeutics, Watertown, USA
- Tiaki Therapeutics, Inc., c/o Dementia Discovery Fund, 201 Washington Street, 39th Floor, Boston, MA, 02108, USA
| | - Michael Marconi
- Department of Molecular Pathology, Massachusetts General Hospital, Boston, USA
- Tiaki Therapeutics, Inc., c/o Dementia Discovery Fund, 201 Washington Street, 39th Floor, Boston, MA, 02108, USA
| | - Christopher Dejesus
- Atalanta Therapeutics, Boston, USA
- Tiaki Therapeutics, Inc., c/o Dementia Discovery Fund, 201 Washington Street, 39th Floor, Boston, MA, 02108, USA
| | - Suzanne Bruhn
- Charcot-Marie-Tooth Association, Glenolden, USA
- Tiaki Therapeutics, Inc., c/o Dementia Discovery Fund, 201 Washington Street, 39th Floor, Boston, MA, 02108, USA
| | - Thomas A Miller
- Walden Biosciences, Cambridge, USA
- Tiaki Therapeutics, Inc., c/o Dementia Discovery Fund, 201 Washington Street, 39th Floor, Boston, MA, 02108, USA
| | - Barbara Tate
- FARA, Homestead, USA
- Tiaki Therapeutics, Inc., c/o Dementia Discovery Fund, 201 Washington Street, 39th Floor, Boston, MA, 02108, USA
| | - Jonathan M Levenson
- FireCyte Therapeutics, Beverly, USA.
- Tiaki Therapeutics, Inc., c/o Dementia Discovery Fund, 201 Washington Street, 39th Floor, Boston, MA, 02108, USA.
| |
Collapse
|
89
|
Korde DS, Humpel C. A Combination of Heavy Metals and Intracellular Pathway Modulators Induces Alzheimer Disease-like Pathologies in Organotypic Brain Slices. Biomolecules 2024; 14:165. [PMID: 38397402 PMCID: PMC10887098 DOI: 10.3390/biom14020165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/17/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that is characterized by amyloid-beta (Aβ) plaques and tau neurofibrillary tangles (NFT). Modelling aspects of AD is challenging due to its complex multifactorial etiology and pathology. The present study aims to establish a cost-effective and rapid method to model the two primary pathologies in organotypic brain slices. Coronal hippocampal brain slices (150 µm) were generated from postnatal (day 8-10) C57BL6 wild-type mice and cultured for 9 weeks. Collagen hydrogels containing either an empty load or a mixture of human Aβ42 and P301S aggregated tau were applied to the slices. The media was further supplemented with various intracellular pathway modulators or heavy metals to augment the appearance of Aβ plaques and tau NFTs, as assessed by immunohistochemistry. Immunoreactivity for Aβ and tau was significantly increased in the ventral areas in slices with a mixture of human Aβ42 and P301S aggregated tau compared to slices with empty hydrogels. Aβ plaque- and tau NFT-like pathologies could be induced independently in slices. Heavy metals (aluminum, lead, cadmium) potently augmented Aβ plaque-like pathology, which developed intracellularly prior to cell death. Intracellular pathway modulators (scopolamine, wortmannin, MHY1485) significantly boosted tau NFT-like pathologies. A combination of nanomolar concentrations of scopolamine, wortmannin, MHY1485, lead, and cadmium in the media strongly increased Aβ plaque- and tau NFT-like immunoreactivity in ventral areas compared to the slices with non-supplemented media. The results highlight that we could harness the potential of the collagen hydrogel-based spreading of human Aβ42 and P301S aggregated tau, along with pharmacological manipulation, to produce pathologies relevant to AD. The results offer a novel ex vivo organotypic slice model to investigate AD pathologies with potential applications for screening drugs or therapies in the future.
Collapse
Affiliation(s)
| | - Christian Humpel
- Laboratory of Psychiatry and Experimental Alzheimer’s Research, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| |
Collapse
|
90
|
Choi GY, Moon E, Choi H, Kweon HS. Changes of synaptic vesicles in three-dimensional synapse models by treatment with umbelliferone in scopolamine-induced hippocampal injury model. Appl Microsc 2024; 54:2. [PMID: 38253782 PMCID: PMC10803702 DOI: 10.1186/s42649-024-00095-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
The neuroprotective effects of umbelliferone (UMB) were visualized in three-dimensional (3D) images on vesicle density changes of organotypic hippocampal slice tissues (OHSCs) induced by scopolamine by high voltage electron microscopy. Observations revealed that the number of vesicles decreased in OHSCs induced by scopolamine, and UMB was found to inhibit scopolamine-induced reduction in vesicles, resulting in an increase in vesicle count. These 3D models provide valuable insight for understanding the increase of synapse vesicles in hippocampal tissues treated with UMB.
Collapse
Affiliation(s)
- Ga-Young Choi
- Center of Research Equipment, Korea Basic Science Institute, Cheongju, 28119, Republic of Korea
| | - Eunyoung Moon
- Center of Research Equipment, Korea Basic Science Institute, Cheongju, 28119, Republic of Korea
| | - Hyosung Choi
- Center of Research Equipment, Korea Basic Science Institute, Cheongju, 28119, Republic of Korea
| | - Hee-Seok Kweon
- Center of Research Equipment, Korea Basic Science Institute, Cheongju, 28119, Republic of Korea.
| |
Collapse
|
91
|
Clavet-Fournier V, Lee C, Wegner W, Brose N, Rhee J, Willig KI. Pre- and postsynaptic nanostructures increase in size and complexity after induction of long-term potentiation. iScience 2024; 27:108679. [PMID: 38213627 PMCID: PMC10783556 DOI: 10.1016/j.isci.2023.108679] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 11/09/2023] [Accepted: 12/05/2023] [Indexed: 01/13/2024] Open
Abstract
Synapses, specialized contact sites between neurons, are the fundamental elements of neuronal information transfer. Synaptic plasticity involves changes in synaptic morphology and the number of neurotransmitter receptors, and is thought to underlie learning and memory. However, it is not clear how these structural and functional changes are connected. We utilized time-lapse super-resolution STED microscopy of organotypic hippocampal brain slices and cultured neurons to visualize structural changes of the synaptic nano-organization of the postsynaptic scaffolding protein PSD95, the presynaptic scaffolding protein Bassoon, and the GluA2 subunit of AMPA receptors by chemically induced long-term potentiation (cLTP) at the level of single synapses. We found that the nano-organization of all three proteins increased in complexity and size after cLTP induction. The increase was largely synchronous, peaking at ∼60 min after stimulation. Therefore, both the size and complexity of individual pre- and post-synaptic nanostructures serve as substrates for tuning and determining synaptic strength.
Collapse
Affiliation(s)
- Valérie Clavet-Fournier
- Group of Optical Nanoscopy in Neuroscience, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Göttingen Graduate Center for Neurosciences, Biophysics, und Molecular Biosciences (GGNB), Göttingen, Germany
| | - ChungKu Lee
- Department of Molecular Neurobiology, Synaptic Physiology Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Waja Wegner
- Group of Optical Nanoscopy in Neuroscience, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Göttingen, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - JeongSeop Rhee
- Department of Molecular Neurobiology, Synaptic Physiology Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Katrin I. Willig
- Group of Optical Nanoscopy in Neuroscience, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
92
|
Claiborne N, Anisimova M, Zito K. Activity-Dependent Stabilization of Nascent Dendritic Spines Requires Nonenzymatic CaMKIIα Function. J Neurosci 2024; 44:e1393222023. [PMID: 38050081 PMCID: PMC10860566 DOI: 10.1523/jneurosci.1393-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 10/18/2023] [Accepted: 11/17/2023] [Indexed: 12/06/2023] Open
Abstract
The outgrowth and stabilization of nascent dendritic spines are crucial processes underlying learning and memory. Most new spines retract shortly after growth; only a small subset is stabilized and integrated into the new circuit connections that support learning. New spine stabilization has been shown to rely upon activity-dependent molecular mechanisms that also contribute to long-term potentiation (LTP) of synaptic strength. Indeed, disruption of the activity-dependent targeting of the kinase CaMKIIα to the GluN2B subunit of the NMDA-type glutamate receptor disrupts both LTP and activity-dependent stabilization of new spines. Yet it is not known which of CaMKIIα's many enzymatic and structural functions are important for new spine stabilization. Here, we used two-photon imaging and photolysis of caged glutamate to monitor the activity-dependent stabilization of new dendritic spines on hippocampal CA1 neurons from mice of both sexes in conditions where CaMKIIα functional and structural interactions were altered. Surprisingly, we found that inhibiting CaMKIIα kinase activity either genetically or pharmacologically did not impair activity-dependent new spine stabilization. In contrast, shRNA knockdown of CaMKIIα abolished activity-dependent new spine stabilization, which was rescued by co-expressing shRNA-resistant full-length CaMKIIα, but not by a truncated monomeric CaMKIIα. Notably, overexpression of phospho-mimetic CaMKIIα-T286D, which exhibits activity-independent targeting to GluN2B, enhanced basal new spine survivorship in the absence of additional glutamatergic stimulation, even when kinase activity was disrupted. Together, our results support a model in which nascent dendritic spine stabilization requires structural and scaffolding interactions mediated by dodecameric CaMKIIα that are independent of its enzymatic activities.
Collapse
Affiliation(s)
- Nicole Claiborne
- Center for Neuroscience, University of California, Davis, California 95618
| | | | - Karen Zito
- Center for Neuroscience, University of California, Davis, California 95618
| |
Collapse
|
93
|
Normoyle KP, Lillis KP, Staley KJ. Synthesis and Characterization of a Novel Concentration-Independent Fluorescent Chloride Indicator, ABP-Dextran, Optimized for Extracellular Chloride Measurement. Biomolecules 2024; 14:77. [PMID: 38254677 PMCID: PMC10813347 DOI: 10.3390/biom14010077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024] Open
Abstract
GABA, the primary inhibitory neurotransmitter, stimulates GABAA receptors (GABAARs) to increase the chloride conductance of the cytosolic membrane. The driving forces for membrane chloride currents are determined by the local differences between intracellular and extracellular chloride concentrations (Cli and Clo, respectively). While several strategies exist for the measurement of Cli, the field lacks tools for non-invasive measurement of Clo. We present the design and development of a fluorescent lifetime imaging (FLIM)-compatible small molecule, N(4-aminobutyl)phenanthridiunium (ABP) with the brightness, spectral features, sensitivity to chloride, and selectivity versus other anions to serve as a useful probe of Clo. ABP can be conjugated to dextran to ensure extracellular compartmentalization, and a second chloride-insensitive counter-label can be added for ratiometric imaging. We validate the utility of this novel sensor series in two sensor concentration-independent modes: FLIM or ratiometric intensity-based imaging.
Collapse
Affiliation(s)
- Kieran P. Normoyle
- Massachusetts General Hospital, Department of Neurology, 55 Fruit Street, Boston, MA 02114, USA; (K.P.N.); (K.P.L.)
- Harvard Medical School, Department of Neurology, 77 Louis Pasteur Avenue, Boston, MA 02115, USA
| | - Kyle P. Lillis
- Massachusetts General Hospital, Department of Neurology, 55 Fruit Street, Boston, MA 02114, USA; (K.P.N.); (K.P.L.)
- Harvard Medical School, Department of Neurology, 77 Louis Pasteur Avenue, Boston, MA 02115, USA
| | - Kevin J. Staley
- Massachusetts General Hospital, Department of Neurology, 55 Fruit Street, Boston, MA 02114, USA; (K.P.N.); (K.P.L.)
- Harvard Medical School, Department of Neurology, 77 Louis Pasteur Avenue, Boston, MA 02115, USA
| |
Collapse
|
94
|
Gellner AK, Reis J, Fiebich BL, Fritsch B. Cx3cr1 deficiency interferes with learning- and direct current stimulation-mediated neuroplasticity of the motor cortex. Eur J Neurosci 2024; 59:177-191. [PMID: 38049944 DOI: 10.1111/ejn.16206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 10/18/2023] [Accepted: 11/12/2023] [Indexed: 12/06/2023]
Abstract
Microglia are essential contributors to synaptic transmission and stability and communicate with neurons via the fractalkine pathway. Transcranial direct current stimulation [(t)DCS], a form of non-invasive electrical brain stimulation, modulates cortical excitability and promotes neuroplasticity, which has been extensively demonstrated in the motor cortex and for motor learning. The role of microglia and their fractalkine receptor CX3CR1 in motor cortical neuroplasticity mediated by DCS or motor learning requires further elucidation. We demonstrate the effects of pharmacological microglial depletion and genetic Cx3cr1 deficiency on the induction of DCS-induced long-term potentiation (DCS-LTP) ex vivo. The relevance of microglia-neuron communication for DCS response and structural neuroplasticity underlying motor learning are assessed via 2-photon in vivo imaging. The behavioural consequences of impaired CX3CR1 signalling are investigated for both gross and fine motor learning. We show that DCS-mediated neuroplasticity in the motor cortex depends on the presence of microglia and is driven in part by CX3CR1 signalling ex vivo and provide the first evidence of microglia interacting with neurons during DCS in vivo. Furthermore, CX3CR1 signalling is required for motor learning and underlying structural neuroplasticity in concert with microglia interaction. Although we have recently demonstrated the microglial response to DCS in vivo, we now provide a link between microglial integrity and neuronal activity for the expression of DCS-dependent neuroplasticity. In addition, we extend the knowledge on the relevance of CX3CR1 signalling for motor learning and structural neuroplasticity. The underlying molecular mechanisms and the potential impact of DCS in rescuing CX3CR1 deficits remain to be addressed in the future.
Collapse
Affiliation(s)
- Anne-Kathrin Gellner
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
- Department of Neurology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute of Physiology II, Medical Faculty, University of Bonn, Bonn, Germany
| | - Janine Reis
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| | - Bernd L Fiebich
- Neurochemistry and Neuroimmunology Research Group, Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Brita Fritsch
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
95
|
Pushkin AN, Kay Y, Herring BE. Protein 4.1N Plays a Cell Type-Specific Role in Hippocampal Glutamatergic Synapse Regulation. J Neurosci 2023; 43:8336-8347. [PMID: 37845032 PMCID: PMC10711697 DOI: 10.1523/jneurosci.0185-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 09/14/2023] [Accepted: 10/02/2023] [Indexed: 10/18/2023] Open
Abstract
Many glutamatergic synapse proteins contain a 4.1N protein binding domain. However, a role for 4.1N in the regulation of glutamatergic neurotransmission has been controversial. Here, we observe significantly higher expression of protein 4.1N in granule neurons of the dentate gyrus (DG granule neurons) compared with other hippocampal regions. We discover that reducing 4.1N expression in rat DG granule neurons of either sex results in a significant reduction in glutamatergic synapse function that is caused by a decrease in the number of glutamatergic synapses. By contrast, we find reduction of 4.1N expression in hippocampal CA1 pyramidal neurons has no impact on basal glutamatergic neurotransmission. We also find 4.1N's C-terminal domain (CTD) to be nonessential to its role in the regulation of glutamatergic synapses of DG granule neurons. Instead, we show that 4.1N's four-point-one, ezrin, radixin, and moesin (FERM) domain is essential for supporting synaptic AMPA receptor (AMPAR) function in these neurons. Altogether, this work demonstrates a novel, cell type-specific role for protein 4.1N in governing glutamatergic synapse function.SIGNIFICANCE STATEMENT Glutamatergic synapses exhibit immense molecular diversity. In comparison to heavily studied Schaffer collateral, CA1 glutamatergic synapses, significantly less is known about perforant path-dentate gyrus (DG) synapses. Our data demonstrate that compromising 4.1N function in CA1 pyramidal neurons produces no alteration in basal glutamatergic synaptic transmission. However, in DG granule neurons, compromising 4.1N function leads to a significant decrease in the strength of glutamatergic neurotransmission at perforant pathway synapses. Together, our data identifies 4.1N as a cell type-specific regulator of synaptic transmission within the hippocampus and reveals a unique molecular program that governs perforant pathway synapse function.
Collapse
Affiliation(s)
- Anna N Pushkin
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California 90089
| | - Yuni Kay
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California 90089
| | - Bruce E Herring
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California 90089
- Department of Biological Sciences, Neurobiology Section, Dornslife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, California 90089
| |
Collapse
|
96
|
Vivi E, Seeholzer LR, Nagumanova A, Di Benedetto B. Early Age- and Sex-Dependent Regulation of Astrocyte-Mediated Glutamatergic Synapse Elimination in the Rat Prefrontal Cortex: Establishing an Organotypic Brain Slice Culture Investigating Tool. Cells 2023; 12:2761. [PMID: 38067189 PMCID: PMC10705965 DOI: 10.3390/cells12232761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/27/2023] [Accepted: 12/01/2023] [Indexed: 12/18/2023] Open
Abstract
Clinical and pre-clinical studies of neuropsychiatric (NP) disorders show altered astrocyte properties and synaptic networks. These are refined during early postnatal developmental (PND) stages. Thus, investigating early brain maturational trajectories is essential to understand NP disorders. However, animal experiments are highly time-/resource-consuming, thereby calling for alternative methodological approaches. The function of MEGF10 in astrocyte-mediated synapse elimination (pruning) is crucial to refine neuronal networks during development and adulthood. To investigate the impact of MEGF10 during PND in the rat prefrontal cortex (PFC) and its putative role in brain disorders, we established and validated an organotypic brain slice culture (OBSC) system. Using Western blot, we characterized the expression of MEGF10 and the synaptic markers synaptophysin and PSD95 in the cortex of developing pups. We then combined immunofluorescent-immunohistochemistry with Imaris-supported 3D analysis to compare age- and sex-dependent astrocyte-mediated pruning within the PFC in pups and OBSCs. We thereby validated this system to investigate age-dependent astrocyte-mediated changes in pruning during PND. However, further optimizations are required to use OBSCs for revealing sex-dependent differences. In conclusion, OBSCs offer a valid alternative to study physiological astrocyte-mediated synaptic remodeling during PND and might be exploited to investigate the pathomechanisms of brain disorders with aberrant synaptic development.
Collapse
Affiliation(s)
- Eugenia Vivi
- Laboratory of Neuro-Glia Pharmacology, Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany; (E.V.); (L.R.S.); (A.N.)
| | - Lea R. Seeholzer
- Laboratory of Neuro-Glia Pharmacology, Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany; (E.V.); (L.R.S.); (A.N.)
| | - Anastasiia Nagumanova
- Laboratory of Neuro-Glia Pharmacology, Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany; (E.V.); (L.R.S.); (A.N.)
| | - Barbara Di Benedetto
- Laboratory of Neuro-Glia Pharmacology, Department of Psychiatry and Psychotherapy, University of Regensburg, 93053 Regensburg, Germany; (E.V.); (L.R.S.); (A.N.)
- Regensburg Center of Neuroscience, University of Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
97
|
Vincent PF, Young ED, Edge AS, Glowatzki E. Auditory Hair Cells and Spiral Ganglion Neurons Regenerate Synapses with Refined Release Properties In Vitro. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.05.561095. [PMID: 38076928 PMCID: PMC10705289 DOI: 10.1101/2023.10.05.561095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Ribbon synapses between inner hair cells (IHCs) and type I spiral ganglion neurons (SGNs) in the inner ear are damaged by noise trauma and with aging, causing 'synaptopathy 'and hearing loss. Co-cultures of neonatal denervated organs of Corti and newly introduced SGNs have been developed to find strategies for improving IHC synapse regeneration, but evidence of the physiological normality of regenerated synapses is missing. This study utilizes IHC optogenetic stimulation and SGN recordings, showing that newly formed IHC synapses are indeed functional, exhibiting glutamatergic excitatory postsynaptic currents. When older organs of Corti were plated, synaptic activity probed by deconvolution, showed more mature release properties, closer to the highly specialized mode of IHC synaptic transmission that is crucial for coding the sound signal. This newly developed functional assessment of regenerated IHC synapses provides a powerful tool for testing approaches to improve synapse regeneration.
Collapse
Affiliation(s)
- Philippe F.Y. Vincent
- The Center for Hearing and Balance, The Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Otolaryngology Head and Neck Surgery, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Eric D. Young
- The Center for Hearing and Balance, The Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Otolaryngology Head and Neck Surgery, The Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Neuroscience, The Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Biomedical Engineering, The Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Albert S.B. Edge
- Department of Otolaryngology, Harvard Medical School, Boston, Massachusetts, USA
- Eaton-Peabody Laboratory, Massachusetts Eye and Ear, Boston, Massachusetts, USA
- Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Elisabeth Glowatzki
- The Center for Hearing and Balance, The Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Otolaryngology Head and Neck Surgery, The Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Neuroscience, The Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
98
|
Moreton N, Puzio M, McCormack J, O'Connor JJ. The effects of prolyl hydroxylase inhibition during and post, hypoxia, oxygen glucose deprivation and oxidative stress, in isolated rat hippocampal slices. Brain Res Bull 2023; 205:110822. [PMID: 37984622 DOI: 10.1016/j.brainresbull.2023.110822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/05/2023] [Accepted: 11/17/2023] [Indexed: 11/22/2023]
Abstract
The contributions of hypoxia and oxidative stress to the pathophysiology of acute ischemic stroke are well established and can lead to disruptions in synaptic signaling. Hypoxia and oxidative stress lead to the neurotoxic overproduction of reactive oxygen species (ROS) and the stabilization of hypoxia inducible factors (HIF). Compounds such as prolyl-4-hydroxylase domain enzyme inhibitors (PHDIs) have been shown to have a preconditioning and neuroprotective effect against ischemic insults such as hypoxia, anoxia, oxygen glucose deprivation (OGD) or H2O2. Therefore, this study explored the effects of two PHDIs, JNJ-42041935 (10 µM) and roxadustat (100 µM) on cell viability using organotypic hippocampal slice cultures. We also assessed the effects of these compounds on synaptic transmission during and post hypoxia, OGD and H2O2 application in isolated rat hippocampal slices using field recording electrophysiological techniques and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit trafficking using immunohistochemistry. Our organotypic data demonstrated a protective role for both inhibitors, where slices had significantly less cell death post anoxia and OGD compared to controls. We also report a distinct modulatory role for both JNJ-42041935 and roxadustat on fEPSP slope post hypoxia and OGD but not H2O2. In addition, we report that application of roxadustat impaired long-term potentiation, but only when applied post-hypoxia. This inhibitory effect was not reversed with co-application of the cyclin-dependent kinase 5 (CDK-5) inhibitor, roscovitine (10 µM), suggesting a CDK-5 independent synaptic AMPAR trafficking mechanism. Both hypoxia and OGD induced a reduction in synaptic AMPA GluA2 subunits, the OGD effect being reversed by prior treatment with both JNJ-42041935 and roxadustat. These results suggest an important role for PHDs in synaptic signaling and plasticity during episodes of ischemic stress.
Collapse
Affiliation(s)
- Niamh Moreton
- UCD School of Biomolecular & Biomedical Science, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Martina Puzio
- UCD School of Biomolecular & Biomedical Science, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Janet McCormack
- UCD Research Pathology Core, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - John J O'Connor
- UCD School of Biomolecular & Biomedical Science, UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
99
|
Hattori R, Hedrick NG, Jain A, Chen S, You H, Hattori M, Choi JH, Lim BK, Yasuda R, Komiyama T. Meta-reinforcement learning via orbitofrontal cortex. Nat Neurosci 2023; 26:2182-2191. [PMID: 37957318 PMCID: PMC10689244 DOI: 10.1038/s41593-023-01485-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 10/06/2023] [Indexed: 11/15/2023]
Abstract
The meta-reinforcement learning (meta-RL) framework, which involves RL over multiple timescales, has been successful in training deep RL models that generalize to new environments. It has been hypothesized that the prefrontal cortex may mediate meta-RL in the brain, but the evidence is scarce. Here we show that the orbitofrontal cortex (OFC) mediates meta-RL. We trained mice and deep RL models on a probabilistic reversal learning task across sessions during which they improved their trial-by-trial RL policy through meta-learning. Ca2+/calmodulin-dependent protein kinase II-dependent synaptic plasticity in OFC was necessary for this meta-learning but not for the within-session trial-by-trial RL in experts. After meta-learning, OFC activity robustly encoded value signals, and OFC inactivation impaired the RL behaviors. Longitudinal tracking of OFC activity revealed that meta-learning gradually shapes population value coding to guide the ongoing behavioral policy. Our results indicate that two distinct RL algorithms with distinct neural mechanisms and timescales coexist in OFC to support adaptive decision-making.
Collapse
Affiliation(s)
- Ryoma Hattori
- Department of Neurobiology, University of California San Diego, La Jolla, CA, USA.
- Center for Neural Circuits and Behavior, University of California San Diego, La Jolla, CA, USA.
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA.
- Halıcıoğlu Data Science Institute, University of California San Diego, La Jolla, CA, USA.
- Department of Neuroscience, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, Jupiter, FL, USA.
| | - Nathan G Hedrick
- Department of Neurobiology, University of California San Diego, La Jolla, CA, USA
- Center for Neural Circuits and Behavior, University of California San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
- Halıcıoğlu Data Science Institute, University of California San Diego, La Jolla, CA, USA
| | - Anant Jain
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Shuqi Chen
- Department of Neurobiology, University of California San Diego, La Jolla, CA, USA
- Center for Neural Circuits and Behavior, University of California San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
- Halıcıoğlu Data Science Institute, University of California San Diego, La Jolla, CA, USA
| | - Hanjia You
- Department of Neurobiology, University of California San Diego, La Jolla, CA, USA
- Center for Neural Circuits and Behavior, University of California San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
- Halıcıoğlu Data Science Institute, University of California San Diego, La Jolla, CA, USA
| | - Mariko Hattori
- Department of Neurobiology, University of California San Diego, La Jolla, CA, USA
- Center for Neural Circuits and Behavior, University of California San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
- Halıcıoğlu Data Science Institute, University of California San Diego, La Jolla, CA, USA
| | - Jun-Hyeok Choi
- Department of Neurobiology, University of California San Diego, La Jolla, CA, USA
| | - Byung Kook Lim
- Department of Neurobiology, University of California San Diego, La Jolla, CA, USA
| | - Ryohei Yasuda
- Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Takaki Komiyama
- Department of Neurobiology, University of California San Diego, La Jolla, CA, USA.
- Center for Neural Circuits and Behavior, University of California San Diego, La Jolla, CA, USA.
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA.
- Halıcıoğlu Data Science Institute, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
100
|
Bagnoli E, Trotier A, McMahon J, Quinlan LR, Biggs M, Pandit A, FitzGerald U. Prodromal Parkinson's disease and the catecholaldehyde hypothesis: Insight from olfactory bulb organotypic cultures. FASEB J 2023; 37:e23272. [PMID: 37997495 DOI: 10.1096/fj.202301253r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/15/2023] [Accepted: 10/10/2023] [Indexed: 11/25/2023]
Abstract
Parkinson's disease (PD) is a progressive, neurodegenerative disorder with an increasing incidence, unknown etiology, and is currently incurable. Advances in understanding the pathological mechanisms at a molecular level have been slow, with little attention focused on the early prodromal phase of the disease. Consequently, the development of early-acting disease-modifying therapies has been hindered. The olfactory bulb (OB), the brain region responsible for initial processing of olfactory information, is particularly affected early in PD at both functional and molecular levels but there is little information on how the cells in this region are affected by disease. Organotypic and primary OB cultures were developed and characterized. These platforms were then used to assess the effects of 3,4-dihydroxyphenylacetylaldehyde (DOPAL), a metabolite of dopamine present in increased levels in post-mortem PD tissue and which is thought to contribute to PD pathogenesis. Our findings showed that DOPAL exposure can recapitulate many aspects of PD pathology. Oxidative stress, depolarization of mitochondrial membranes, and neurodegeneration were all induced by DOPAL addition, as were measured transcriptomic changes consistent with those reported in PD clinical studies. These olfactory models of prodromal disease lend credence to the catecholaldehyde hypothesis of PD and provide insight into the mechanisms by which the OB may be involved in disease progression.
Collapse
Affiliation(s)
- Enrico Bagnoli
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
- Galway Neuroscience Centre, University of Galway, Galway, Ireland
| | - Alexandre Trotier
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
- Galway Neuroscience Centre, University of Galway, Galway, Ireland
| | - Jill McMahon
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
- Galway Neuroscience Centre, University of Galway, Galway, Ireland
| | - Leo R Quinlan
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
- Physiology, School of Medicine, Galway, Ireland
| | - Manus Biggs
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
- Galway Neuroscience Centre, University of Galway, Galway, Ireland
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
- Galway Neuroscience Centre, University of Galway, Galway, Ireland
| | - Una FitzGerald
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
- Galway Neuroscience Centre, University of Galway, Galway, Ireland
| |
Collapse
|