1
|
Bosworth AP, Contreras M, Sancho L, Salas IH, Paumier A, Novak SW, Manor U, Allen NJ. Astrocyte glypican 5 regulates synapse maturation and stabilization. Cell Rep 2025; 44:115374. [PMID: 40048429 PMCID: PMC12013928 DOI: 10.1016/j.celrep.2025.115374] [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: 02/21/2023] [Revised: 11/28/2024] [Accepted: 02/10/2025] [Indexed: 03/29/2025] Open
Abstract
The maturation and stabilization of appropriate synaptic connections is a vital step in neural circuit development; however, the molecular signals underlying these processes are not fully understood. We show that astrocytes, through production of glypican 5 (GPC5), are required for maturation and refinement of synapses in the mouse cortex during the critical period. In the absence of astrocyte GPC5, thalamocortical synapses show structural immaturity, including smaller presynaptic terminals, decreased postsynaptic density area, and presence of more postsynaptic partners at multisynaptic connections. This structural immaturity is accompanied by a delay in developmental incorporation of GLUA2-containing AMPARs at intracortical synapses. The functional impact of this is that mice lacking astrocyte GPC5 exhibit increased levels of ocular dominance plasticity in adulthood. This demonstrates that astrocyte GPC5 is necessary for maturation and stabilization of synaptic connections, which has implications for disorders with altered synaptic function where GPC5 levels are altered, including Alzheimer's disease and frontotemporal dementia.
Collapse
Affiliation(s)
- Alexandra P Bosworth
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Rd., La Jolla, CA 92037, USA; Neurosciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Minerva Contreras
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Rd., La Jolla, CA 92037, USA; Neurosciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Laura Sancho
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Rd., La Jolla, CA 92037, USA
| | - Isabel H Salas
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Rd., La Jolla, CA 92037, USA
| | - Adrien Paumier
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Rd., La Jolla, CA 92037, USA
| | - Sammy Weiser Novak
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, 10010 North Torrey Pines Rd., La Jolla, CA 92037, USA
| | - Uri Manor
- Waitt Advanced Biophotonics Center, The Salk Institute for Biological Studies, 10010 North Torrey Pines Rd., La Jolla, CA 92037, USA; Department of Cell & Developmental Biology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nicola J Allen
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Rd., La Jolla, CA 92037, USA.
| |
Collapse
|
2
|
Kerckhof P, Ambrocio GPL, Beeckmans H, Kaes J, Geudens V, Bos S, Willems L, Vermaut A, Vermant M, Goos T, De Fays C, Aversa L, Mohamady Y, Vanstapel A, Orlitová M, Van Slambrouck J, Jin X, Varghese V, Josipovic I, Boone MN, Dupont LJ, Weynand B, Dubbeldam A, Van Raemdonck DE, Ceulemans LJ, Gayan-Ramirez G, De Sadeleer LJ, McDonough JE, Vanaudenaerde BM, Vos R. Ventilatory capacity in CLAD is driven by dysfunctional airway structure. EBioMedicine 2024; 101:105030. [PMID: 38394744 PMCID: PMC10897920 DOI: 10.1016/j.ebiom.2024.105030] [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: 11/15/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Chronic lung allograft dysfunction (CLAD) encompasses three main phenotypes: bronchiolitis obliterans syndrome (BOS), restrictive allograft syndrome (RAS) and a Mixed phenotype combining both pathologies. How the airway structure in its entirety is affected in these phenotypes is still poorly understood. METHODS A detailed analysis of airway morphometry was applied to gain insights on the effects of airway remodelling on the distribution of alveolar ventilation in end-stage CLAD. Ex vivo whole lung μCT and tissue-core μCT scanning of six control, six BOS, three RAS and three Mixed explant lung grafts (9 male, 9 female, 2014-2021, Leuven, Belgium) were used for digital airway reconstruction and calculation of airway dimensions in relation to luminal obstructions. FINDINGS BOS and Mixed explants demonstrated airway obstructions of proximal bronchioles (starting at generation five), while RAS explants particularly had airway obstructions in the most distal bronchioles (generation >12). In BOS and Mixed explants 76% and 84% of bronchioles were obstructed, respectively, while this was 22% in RAS. Bronchiolar obstructions were mainly caused by lymphocytic inflammation of the airway wall or fibrotic remodelling, i.e. constrictive bronchiolitis. Proximal bronchiolectasis and imbalance in distal lung ventilation were present in all CLAD phenotypes and explain poor lung function and deterioration of specific lung function parameters. INTERPRETATION Alterations in the structure of conducting bronchioles revealed CLAD to affect alveolar ventilatory distribution in a regional fashion. The significance of various obstructions, particularly those associated with mucus, is highlighted. FUNDING This research was funded with the National research fund Flanders (G060322N), received by R.V.
Collapse
Affiliation(s)
- Pieterjan Kerckhof
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Gene P L Ambrocio
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Division of Pulmonary Medicine, Department of Internal Medicine, University of the Philippines - Philippine General Hospital, Manilla, The Philippines
| | - Hanne Beeckmans
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Janne Kaes
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Vincent Geudens
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Saskia Bos
- Newcastle University, Translational and Clinical Research Institute, Newcastle upon Tyne, United Kingdom
| | - Lynn Willems
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Astrid Vermaut
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Marie Vermant
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Tinne Goos
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Charlotte De Fays
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université Catholique de Louvain, Brussels, Belgium
| | - Lucia Aversa
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Yousry Mohamady
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Arno Vanstapel
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | | | - Jan Van Slambrouck
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Xin Jin
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Vimi Varghese
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Department of Heart and Lung Transplant, Yashoda Hospitals, Hyderabad, India
| | - Iván Josipovic
- Department of Physics and Astronomy, UGCT, Radiation Physics, Ghent University, Gent, Belgium
| | - Matthieu N Boone
- Department of Physics and Astronomy, UGCT, Radiation Physics, Ghent University, Gent, Belgium
| | - Lieven J Dupont
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Birgit Weynand
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Adriana Dubbeldam
- Department of Radiology, University Hospitals Leuven, Leuven, Belgium
| | | | - Laurens J Ceulemans
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Ghislaine Gayan-Ramirez
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Laurens J De Sadeleer
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium; Cell Circuits in Systems Medicine of Lung Disease (Schiller Lab), Institute of Lung Health and Immunity (LHI) / Comprehensive Pneumology Centre (CPC), German Centre for Lung Research, Helmholtz Zentrum München, München, Germany
| | - John E McDonough
- Department of Medicine, McMaster University, Firestone Institute of Respiratory Health, Hamilton, Canada
| | - Bart M Vanaudenaerde
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department of CHROMETA, KU Leuven, Leuven, Belgium
| | - Robin Vos
- Department of Respiratory Diseases, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
3
|
Li W, Chen R, Feng L, Dang X, Liu J, Chen T, Yang J, Su X, Lv L, Li T, Zhang Z, Luo XJ. Genome-wide meta-analysis, functional genomics and integrative analyses implicate new risk genes and therapeutic targets for anxiety disorders. Nat Hum Behav 2024; 8:361-379. [PMID: 37945807 DOI: 10.1038/s41562-023-01746-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 10/04/2023] [Indexed: 11/12/2023]
Abstract
Anxiety disorders are the most prevalent mental disorders. However, the genetic etiology of anxiety disorders remains largely unknown. Here we conducted a genome-wide meta-analysis on anxiety disorders by including 74,973 (28,392 proxy) cases and 400,243 (146,771 proxy) controls. We identified 14 risk loci, including 10 new associations near CNTNAP5, MAP2, RAB9BP1, BTN1A1, PRR16, PCLO, PTPRD, FARP1, CDH2 and RAB27B. Functional genomics and fine-mapping pinpointed the potential causal variants, and expression quantitative trait loci analysis revealed the potential target genes regulated by the risk variants. Integrative analyses, including transcriptome-wide association study, proteome-wide association study and colocalization analyses, prioritized potential causal genes (including CTNND1 and RAB27B). Evidence from multiple analyses revealed possibly causal genes, including RAB27B, BTN3A2, PCLO and CTNND1. Finally, we showed that Ctnnd1 knockdown affected dendritic spine density and resulted in anxiety-like behaviours in mice, revealing the potential role of CTNND1 in anxiety disorders. Our study identified new risk loci, potential causal variants and genes for anxiety disorders, providing insights into the genetic architecture of anxiety disorders and potential therapeutic targets.
Collapse
Affiliation(s)
- Wenqiang Li
- Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Rui Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Laipeng Feng
- Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Xinglun Dang
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, China
| | - Jiewei Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Tengfei Chen
- Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jinfeng Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Xi Su
- Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Luxian Lv
- Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Tao Li
- Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhijun Zhang
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, China
- Department of Neurology, Affiliated Zhongda Hospital, Southeast University, Nanjing, China
- Department of Mental Health and Public Health, Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xiong-Jian Luo
- Department of Psychosomatics and Psychiatry, Zhongda Hospital, School of Medicine, Advanced Institute for Life and Health, Southeast University, Nanjing, China.
- Department of Neurology, Affiliated Zhongda Hospital, Southeast University, Nanjing, China.
| |
Collapse
|
4
|
Ramos-Brossier M, Romeo-Guitart D, Lanté F, Boitez V, Mailliet F, Saha S, Rivagorda M, Siopi E, Nemazanyy I, Leroy C, Moriceau S, Beck-Cormier S, Codogno P, Buisson A, Beck L, Friedlander G, Oury F. Slc20a1 and Slc20a2 regulate neuronal plasticity and cognition independently of their phosphate transport ability. Cell Death Dis 2024; 15:20. [PMID: 38195526 PMCID: PMC10776841 DOI: 10.1038/s41419-023-06292-z] [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: 04/03/2023] [Revised: 11/01/2023] [Accepted: 11/13/2023] [Indexed: 01/11/2024]
Abstract
In recent years, primary familial brain calcification (PFBC), a rare neurological disease characterized by a wide spectrum of cognitive disorders, has been associated to mutations in the sodium (Na)-Phosphate (Pi) co-transporter SLC20A2. However, the functional roles of the Na-Pi co-transporters in the brain remain still largely elusive. Here we show that Slc20a1 (PiT-1) and Slc20a2 (PiT-2) are the most abundant Na-Pi co-transporters expressed in the brain and are involved in the control of hippocampal-dependent learning and memory. We reveal that Slc20a1 and Slc20a2 are differentially distributed in the hippocampus and associated with independent gene clusters, suggesting that they influence cognition by different mechanisms. Accordingly, using a combination of molecular, electrophysiological and behavioral analyses, we show that while PiT-2 favors hippocampal neuronal branching and survival, PiT-1 promotes synaptic plasticity. The latter relies on a likely Otoferlin-dependent regulation of synaptic vesicle trafficking, which impacts the GABAergic system. These results provide the first demonstration that Na-Pi co-transporters play key albeit distinct roles in the hippocampus pertaining to the control of neuronal plasticity and cognition. These findings could provide the foundation for the development of novel effective therapies for PFBC and cognitive disorders.
Collapse
Affiliation(s)
- Mariana Ramos-Brossier
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Team 8, F-75015, Paris, France.
| | - David Romeo-Guitart
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Team 8, F-75015, Paris, France
| | - Fabien Lanté
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Valérie Boitez
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Team 8, F-75015, Paris, France
| | - François Mailliet
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Team 8, F-75015, Paris, France
| | - Soham Saha
- Institut Pasteur, Perception & Memory Unit, F-75015, Paris, France
- MedInsights, 6 rue de l'église, F-02810, Veuilly la Poterie, France
| | - Manon Rivagorda
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Team 8, F-75015, Paris, France
| | - Eleni Siopi
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Team 8, F-75015, Paris, France
| | - Ivan Nemazanyy
- Platform for Metabolic Analyses, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR, 3633, Paris, France
| | - Christine Leroy
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Team 6, F-75015, Paris, France
| | - Stéphanie Moriceau
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Team 8, F-75015, Paris, France
- Platform for Neurobehavioural and metabolism, Structure Fédérative de Recherche Necker, INSERM, US24/CNRS UAR, 3633, Paris, France
- Institute of Genetic Diseases, Imagine, 75015, Paris, France
| | - Sarah Beck-Cormier
- Nantes Université, CNRS, Inserm, l'Institut du Thorax, F-44000, Nantes, France
| | - Patrice Codogno
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Team 6, F-75015, Paris, France
| | - Alain Buisson
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Laurent Beck
- Nantes Université, CNRS, Inserm, l'Institut du Thorax, F-44000, Nantes, France.
| | - Gérard Friedlander
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Team 6, F-75015, Paris, France.
| | - Franck Oury
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Team 8, F-75015, Paris, France.
| |
Collapse
|
5
|
Rowland C, Moslehi S, Smith JH, Harland B, Dalrymple-Alford J, Taylor RP. Fractal Resonance: Can Fractal Geometry Be Used to Optimize the Connectivity of Neurons to Artificial Implants? ADVANCES IN NEUROBIOLOGY 2024; 36:877-906. [PMID: 38468068 DOI: 10.1007/978-3-031-47606-8_44] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
In parallel to medical applications, exploring how neurons interact with the artificial interface of implants in the human body can be used to learn about their fundamental behavior. For both fundamental and applied research, it is important to determine the conditions that encourage neurons to maintain their natural behavior during these interactions. Whereas previous biocompatibility studies have focused on the material properties of the neuron-implant interface, here we discuss the concept of fractal resonance - the possibility that favorable connectivity properties might emerge by matching the fractal geometry of the implant surface to that of the neurons.To investigate fractal resonance, we first determine the degree to which neurons are fractal and the impact of this fractality on their functionality. By analyzing three-dimensional images of rat hippocampal neurons, we find that the way their dendrites fork and weave through space is important for generating their fractal-like behavior. By modeling variations in neuron connectivity along with the associated energetic and material costs, we highlight how the neurons' fractal dimension optimizes these constraints. To simulate neuron interactions with implant interfaces, we distort the neuron models away from their natural form by modifying the dendrites' fork and weaving patterns. We find that small deviations can induce large changes in fractal dimension, causing the balance between connectivity and cost to deteriorate rapidly. We propose that implant surfaces should be patterned to match the fractal dimension of the neurons, allowing them to maintain their natural functionality as they interact with the implant.
Collapse
Affiliation(s)
- C Rowland
- Physics Department, University of Oregon, Eugene, OR, USA
| | - S Moslehi
- Physics Department, University of Oregon, Eugene, OR, USA
| | - J H Smith
- Physics Department, University of Oregon, Eugene, OR, USA
| | - B Harland
- School of Pharmacy, University of Auckland, Auckland, New Zealand
| | - J Dalrymple-Alford
- School of Psychology, Speech and Hearing, University of Canterbury, Christchurch, New Zealand
| | - R P Taylor
- Physics Department, University of Oregon, Eugene, OR, USA.
| |
Collapse
|
6
|
Gregory BA, Thompson CH, Salatino JW, Railing MJ, Zimmerman AF, Gupta B, Williams K, Beatty JA, Cox CL, Purcell EK. Structural and functional changes of deep layer pyramidal neurons surrounding microelectrode arrays implanted in rat motor cortex. Acta Biomater 2023; 168:429-439. [PMID: 37499727 PMCID: PMC10441615 DOI: 10.1016/j.actbio.2023.07.027] [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: 03/14/2023] [Revised: 06/25/2023] [Accepted: 07/18/2023] [Indexed: 07/29/2023]
Abstract
Devices capable of recording or stimulating neuronal signals have created new opportunities to understand normal physiology and treat sources of pathology in the brain. However, it is possible that the tissue response to implanted electrodes may influence the nature of the signals detected or stimulated. In this study, we characterized structural and functional changes in deep layer pyramidal neurons surrounding silicon or polyimide-based electrodes implanted in the motor cortex of rats. Devices were captured in 300 µm-thick tissue slices collected at the 1 or 6 week time point post-implantation, and individual neurons were assessed using a combination of whole-cell electrophysiology and 2-photon imaging. We observed disrupted dendritic arbors and a significant reduction in spine densities in neurons surrounding devices. These effects were accompanied by a decrease in the frequency of spontaneous excitatory post-synaptic currents, a reduction in sag amplitude, an increase in spike frequency adaptation, and an increase in filopodia density. We hypothesize that the effects observed in this study may contribute to the signal loss and instability that often accompany chronically implanted electrodes. STATEMENT OF SIGNIFICANCE: Implanted electrodes in the brain can be used to treat sources of pathology and understand normal physiology by recording or stimulating electrical signals generated by local neurons. However, a foreign body response following implantation undermines the performance of these devices. While several studies have investigated the biological mechanisms of device-tissue interactions through histology, transcriptomics, and imaging, our study is the first to directly interrogate effects on the function of neurons surrounding electrodes using single-cell electrophysiology. Additionally, we provide new, detailed assessments of the impacts of electrodes on the dendritic structure and spine morphology of neurons, and we assess effects for both traditional (silicon) and newer polymer electrode materials. These results reveal new potential mechanisms of electrode-tissue interactions.
Collapse
Affiliation(s)
| | - Cort H Thompson
- Department of Biomedical Engineering, Michigan State University, United States
| | - Joseph W Salatino
- Department of Biomedical Engineering, Michigan State University, United States
| | - Mia J Railing
- Department of Physiology, Michigan State University, United States
| | | | - Bhavna Gupta
- Neuroscience Program, Michigan State University, United States
| | - Kathleen Williams
- Department of Biomedical Engineering, Michigan State University, United States
| | - Joseph A Beatty
- Department of Physiology, Michigan State University, United States; Neuroscience Program, Michigan State University, United States
| | - Charles L Cox
- Department of Physiology, Michigan State University, United States; Neuroscience Program, Michigan State University, United States
| | - Erin K Purcell
- Department of Biomedical Engineering, Michigan State University, United States; Neuroscience Program, Michigan State University, United States; Department of Electrical and Computer Engineering, Michigan State University, East Lansing, MI, United States.
| |
Collapse
|
7
|
Yin MY, Guo L, Zhao LJ, Zhang C, Liu WP, Zhang CY, Huo JH, Wang L, Li SW, Zheng CB, Xiao X, Li M, Wang C, Chang H. Reduced Vrk2 expression is associated with higher risk of depression in humans and mediates depressive-like behaviors in mice. BMC Med 2023; 21:256. [PMID: 37452335 PMCID: PMC10349461 DOI: 10.1186/s12916-023-02945-0] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 06/15/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND Genome-wide association studies (GWAS) have reported single-nucleotide polymorphisms (SNPs) in the VRK serine/threonine kinase 2 gene (VRK2) showing genome-wide significant associations with major depression, but the regulation effect of the risk SNPs on VRK2 as well as their roles in the illness are yet to be elucidated. METHODS Based on the summary statistics of major depression GWAS, we conducted population genetic analyses, epigenome bioinformatics analyses, dual luciferase reporter assays, and expression quantitative trait loci (eQTL) analyses to identify the functional SNPs regulating VRK2; we also carried out behavioral assessments, dendritic spine morphological analyses, and phosphorylated 4D-label-free quantitative proteomics analyses in mice with Vrk2 repression. RESULTS We identified a SNP rs2678907 located in the 5' upstream of VRK2 gene exhibiting large spatial overlap with enhancer regulatory marks in human neural cells and brain tissues. Using luciferase reporter gene assays and eQTL analyses, the depression risk allele of rs2678907 decreased enhancer activities and predicted lower VRK2 mRNA expression, which is consistent with the observations of reduced VRK2 level in the patients with major depression compared with controls. Notably, Vrk2-/- mice exhibited depressive-like behaviors compared to Vrk2+/+ mice and specifically repressing Vrk2 in the ventral hippocampus using adeno-associated virus (AAV) lead to consistent and even stronger depressive-like behaviors in mice. Compared with Vrk2+/+ mice, the density of mushroom and thin spines in the ventral hippocampus was significantly altered in Vrk2-/- mice, which is in line with the phosphoproteomic analyses showing dysregulated synapse-associated proteins and pathways in Vrk2-/- mice. CONCLUSIONS Vrk2 deficiency mice showed behavioral abnormalities that mimic human depressive phenotypes, which may serve as a useful murine model for studying the pathophysiology of depression.
Collapse
Affiliation(s)
- Mei-Yu Yin
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Lei Guo
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Li-Juan Zhao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Chen Zhang
- Clinical Research Center & Division of Mood Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China.
| | - Wei-Peng Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Chu-Yi Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Jin-Hua Huo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Lu Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Shi-Wu Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Chang-Bo Zheng
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, Yunnan, China
| | - Xiao Xiao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China.
| | - Chuang Wang
- Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China.
- School of Basic Medical Science, Health Science Center, Ningbo University, Ningbo, Zhejiang, China.
| | - Hong Chang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| |
Collapse
|
8
|
McMahon A, Andrews R, Groves D, Ghani SV, Cordes T, Kapanidis AN, Robb NC. High-throughput super-resolution analysis of influenza virus pleomorphism reveals insights into viral spatial organization. PLoS Pathog 2023; 19:e1011484. [PMID: 37390113 DOI: 10.1371/journal.ppat.1011484] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 06/14/2023] [Indexed: 07/02/2023] Open
Abstract
Many viruses form highly pleomorphic particles. In influenza, virion structure is of interest not only in the context of virus assembly, but also because pleomorphic variations may correlate with infectivity and pathogenicity. We have used fluorescence super-resolution microscopy combined with a rapid automated analysis pipeline, a method well-suited to the study of large numbers of pleomorphic structures, to image many thousands of individual influenza virions; gaining information on their size, morphology and the distribution of membrane-embedded and internal proteins. We observed broad phenotypic variability in filament size, and Fourier transform analysis of super resolution images demonstrated no generalized common spatial frequency patterning of HA or NA on the virion surface, suggesting a model of virus particle assembly where the release of progeny filaments from cells occurs in a stochastic way. We also showed that viral RNP complexes are located preferentially within Archetti bodies when these were observed at filament ends, suggesting that these structures may play a role in virus transmission. Our approach therefore offers exciting new insights into influenza virus morphology and represents a powerful technique that is easily extendable to the study of pleomorphism in other pathogenic viruses.
Collapse
Affiliation(s)
- Andrew McMahon
- Biological Physics, Department of Physics, University of Oxford, Oxford, United Kingdom
- Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, Oxford, United Kingdom
- Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Rebecca Andrews
- Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Danielle Groves
- Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Sohail V Ghani
- Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Thorben Cordes
- Physical and Synthetic Biology, Faculty of Biology, Ludwig-Maximilians-Universität München, Großhadernerstr, Planegg-Martinsried, Germany
| | - Achillefs N Kapanidis
- Biological Physics, Department of Physics, University of Oxford, Oxford, United Kingdom
- Kavli Institute for Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, University of Oxford, Oxford, United Kingdom
| | - Nicole C Robb
- Biological Physics, Department of Physics, University of Oxford, Oxford, United Kingdom
- Warwick Medical School, University of Warwick, Coventry, United Kingdom
| |
Collapse
|
9
|
Knaus LS, Basilico B, Malzl D, Gerykova Bujalkova M, Smogavec M, Schwarz LA, Gorkiewicz S, Amberg N, Pauler FM, Knittl-Frank C, Tassinari M, Maulide N, Rülicke T, Menche J, Hippenmeyer S, Novarino G. Large neutral amino acid levels tune perinatal neuronal excitability and survival. Cell 2023; 186:1950-1967.e25. [PMID: 36996814 DOI: 10.1016/j.cell.2023.02.037] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 02/03/2023] [Accepted: 02/23/2023] [Indexed: 03/31/2023]
Abstract
Little is known about the critical metabolic changes that neural cells have to undergo during development and how temporary shifts in this program can influence brain circuitries and behavior. Inspired by the discovery that mutations in SLC7A5, a transporter of metabolically essential large neutral amino acids (LNAAs), lead to autism, we employed metabolomic profiling to study the metabolic states of the cerebral cortex across different developmental stages. We found that the forebrain undergoes significant metabolic remodeling throughout development, with certain groups of metabolites showing stage-specific changes, but what are the consequences of perturbing this metabolic program? By manipulating Slc7a5 expression in neural cells, we found that the metabolism of LNAAs and lipids are interconnected in the cortex. Deletion of Slc7a5 in neurons affects the postnatal metabolic state, leading to a shift in lipid metabolism. Additionally, it causes stage- and cell-type-specific alterations in neuronal activity patterns, resulting in a long-term circuit dysfunction.
Collapse
Affiliation(s)
- Lisa S Knaus
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria
| | - Bernadette Basilico
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria
| | - Daniel Malzl
- Max Perutz Labs, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
| | - Maria Gerykova Bujalkova
- Institute of Medical Genetics, Medical University of Vienna, Währinger Straße 10, 1090 Vienna, Austria
| | - Mateja Smogavec
- Institute of Medical Genetics, Medical University of Vienna, Währinger Straße 10, 1090 Vienna, Austria
| | - Lena A Schwarz
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria
| | - Sarah Gorkiewicz
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria
| | - Nicole Amberg
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria
| | - Florian M Pauler
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria
| | - Christian Knittl-Frank
- Institute of Organic Chemistry, University of Vienna, Währinger Strasse 38, 1090 Vienna, Austria
| | - Marianna Tassinari
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria
| | - Nuno Maulide
- Institute of Organic Chemistry, University of Vienna, Währinger Strasse 38, 1090 Vienna, Austria; University of Vienna, Research Platform NeGeMac, Währinger Strasse 38, 1090 Vienna, Austria
| | - Thomas Rülicke
- University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria
| | - Jörg Menche
- Max Perutz Labs, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
| | - Simon Hippenmeyer
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria
| | - Gaia Novarino
- Institute of Science and Technology Austria (ISTA), Am Campus 1, 3400 Klosterneuburg, Austria.
| |
Collapse
|
10
|
Sharma A, Sharma N, Singh S, Dua K. Review on theranostic and neuroprotective applications of nanotechnology in multiple sclerosis. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
11
|
Liu Y, Wang G, Ascoli GA, Zhou J, Liu L. Neuron tracing from light microscopy images: automation, deep learning and bench testing. Bioinformatics 2022; 38:5329-5339. [PMID: 36303315 PMCID: PMC9750132 DOI: 10.1093/bioinformatics/btac712] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/19/2022] [Accepted: 10/26/2022] [Indexed: 12/24/2022] Open
Abstract
MOTIVATION Large-scale neuronal morphologies are essential to neuronal typing, connectivity characterization and brain modeling. It is widely accepted that automation is critical to the production of neuronal morphology. Despite previous survey papers about neuron tracing from light microscopy data in the last decade, thanks to the rapid development of the field, there is a need to update recent progress in a review focusing on new methods and remarkable applications. RESULTS This review outlines neuron tracing in various scenarios with the goal to help the community understand and navigate tools and resources. We describe the status, examples and accessibility of automatic neuron tracing. We survey recent advances of the increasingly popular deep-learning enhanced methods. We highlight the semi-automatic methods for single neuron tracing of mammalian whole brains as well as the resulting datasets, each containing thousands of full neuron morphologies. Finally, we exemplify the commonly used datasets and metrics for neuron tracing bench testing.
Collapse
Affiliation(s)
- Yufeng Liu
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Gaoyu Wang
- School of Computer Science and Engineering, Southeast University, Nanjing, China
| | - Giorgio A Ascoli
- Center for Neural Informatics, Structures, & Plasticity, Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, USA
| | - Jiangning Zhou
- Institute of Brain Science, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lijuan Liu
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| |
Collapse
|
12
|
Bhattacharjee S, Lottes EN, Nanda S, Golshir A, Patel AA, Ascoli GA, Cox DN. PP2A phosphatase regulates cell-type specific cytoskeletal organization to drive dendrite diversity. Front Mol Neurosci 2022; 15:926567. [PMID: 36452406 PMCID: PMC9702092 DOI: 10.3389/fnmol.2022.926567] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 10/27/2022] [Indexed: 11/15/2022] Open
Abstract
Uncovering molecular mechanisms regulating dendritic diversification is essential to understanding the formation and modulation of functional neural circuitry. Transcription factors play critical roles in promoting dendritic diversity and here, we identify PP2A phosphatase function as a downstream effector of Cut-mediated transcriptional regulation of dendrite development. Mutant analyses of the PP2A catalytic subunit (mts) or the scaffolding subunit (PP2A-29B) reveal cell-type specific regulatory effects with the PP2A complex required to promote dendritic growth and branching in Drosophila Class IV (CIV) multidendritic (md) neurons, whereas in Class I (CI) md neurons, PP2A functions in restricting dendritic arborization. Cytoskeletal analyses reveal requirements for Mts in regulating microtubule stability/polarity and F-actin organization/dynamics. In CIV neurons, mts knockdown leads to reductions in dendritic localization of organelles including mitochondria and satellite Golgi outposts, while CI neurons show increased Golgi outpost trafficking along the dendritic arbor. Further, mts mutant neurons exhibit defects in neuronal polarity/compartmentalization. Finally, genetic interaction analyses suggest β-tubulin subunit 85D is a common PP2A target in CI and CIV neurons, while FoxO is a putative target in CI neurons.
Collapse
Affiliation(s)
| | - Erin N. Lottes
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | - Sumit Nanda
- Center for Neural Informatics, Structures, and Plasticity, Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, United States
| | - Andre Golshir
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | - Atit A. Patel
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | - Giorgio A. Ascoli
- Center for Neural Informatics, Structures, and Plasticity, Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, United States
| | - Daniel N. Cox
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
13
|
Guerra KTK, Renner J, Vásquez CE, Rasia‐Filho AA. Human cortical amygdala dendrites and spines morphology under open‐source three‐dimensional reconstruction procedures. J Comp Neurol 2022; 531:344-365. [PMID: 36355397 DOI: 10.1002/cne.25430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/05/2022] [Accepted: 10/14/2022] [Indexed: 11/12/2022]
Abstract
Visualizing nerve cells has been fundamental for the systematic description of brain structure and function in humans and other species. Different approaches aimed to unravel the morphological features of neuron types and diversity. The inherent complexity of the human nervous tissue and the need for proper histological processing have made studying human dendrites and spines challenging in postmortem samples. In this study, we used Golgi data and open-source software for 3D image reconstruction of human neurons from the cortical amygdaloid nucleus to show different dendrites and pleomorphic spines at different angles. Procedures required minimal equipment and generated high-quality images for differently shaped cells. We used the "single-section" Golgi method adapted for the human brain to engender 3D reconstructed images of the neuronal cell body and the dendritic ramification by adopting a neuronal tracing procedure. In addition, we elaborated 3D reconstructions to visualize heterogeneous dendritic spines using a supervised machine learning-based algorithm for image segmentation. These tools provided an additional upgrade and enhanced visual display of information related to the spatial orientation of dendritic branches and for dendritic spines of varied sizes and shapes in these human subcortical neurons. This same approach can be adapted for other techniques, areas of the central or peripheral nervous system, and comparative analysis between species.
Collapse
Affiliation(s)
- Kétlyn T. Knak Guerra
- Graduate Program in Neuroscience Universidade Federal do Rio Grande do Sul Porto Alegre Brazil
| | - Josué Renner
- Department of Basic Sciences/Physiology Universidade Federal de Ciências da Saúde de Porto Alegre Porto Alegre Brazil
- Graduate Program in Biosciences Universidade Federal de Ciências da Saúde de Porto Alegre Porto Alegre Brazil
| | - Carlos E. Vásquez
- Graduate Program in Neuroscience Universidade Federal do Rio Grande do Sul Porto Alegre Brazil
| | - Alberto A. Rasia‐Filho
- Graduate Program in Neuroscience Universidade Federal do Rio Grande do Sul Porto Alegre Brazil
- Department of Basic Sciences/Physiology Universidade Federal de Ciências da Saúde de Porto Alegre Porto Alegre Brazil
- Graduate Program in Biosciences Universidade Federal de Ciências da Saúde de Porto Alegre Porto Alegre Brazil
| |
Collapse
|
14
|
Chen R, Yang Z, Liu J, Cai X, Huo Y, Zhang Z, Li M, Chang H, Luo XJ. Functional genomic analysis delineates regulatory mechanisms of GWAS-identified bipolar disorder risk variants. Genome Med 2022; 14:53. [PMID: 35590387 PMCID: PMC9121601 DOI: 10.1186/s13073-022-01057-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 05/11/2022] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Genome-wide association studies (GWASs) have identified multiple risk loci for bipolar disorder (BD). However, pinpointing functional (or causal) variants in the reported risk loci and elucidating their regulatory mechanisms remain challenging. METHODS We first integrated chromatin immunoprecipitation sequencing (ChIP-Seq) data from human brain tissues (or neuronal cell lines) and position weight matrix (PWM) data to identify functional single-nucleotide polymorphisms (SNPs). Then, we verified the regulatory effects of these transcription factor (TF) binding-disrupting SNPs (hereafter referred to as "functional SNPs") through a series of experiments, including reporter gene assays, allele-specific expression (ASE) analysis, TF knockdown, CRISPR/Cas9-mediated genome editing, and expression quantitative trait loci (eQTL) analysis. Finally, we overexpressed PACS1 (whose expression was most significantly associated with the identified functional SNPs rs10896081 and rs3862386) in mouse primary cortical neurons to investigate if PACS1 affects dendritic spine density. RESULTS We identified 16 functional SNPs (in 9 risk loci); these functional SNPs disrupted the binding of 7 TFs, for example, CTCF and REST binding was frequently disrupted. We then identified the potential target genes whose expression in the human brain was regulated by these functional SNPs through eQTL analysis. Of note, we showed dysregulation of some target genes of the identified TF binding-disrupting SNPs in BD patients compared with controls, and overexpression of PACS1 reduced the density of dendritic spines, revealing the possible biological mechanisms of these functional SNPs in BD. CONCLUSIONS Our study identifies functional SNPs in some reported risk loci and sheds light on the regulatory mechanisms of BD risk variants. Further functional characterization and mechanistic studies of these functional SNPs and candidate genes will help to elucidate BD pathogenesis and develop new therapeutic approaches and drugs.
Collapse
Affiliation(s)
- Rui Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Zhihui Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Jiewei Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Xin Cai
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, 650204, China
| | - Yongxia Huo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China
| | - Zhijun Zhang
- Department of Neurology, Affiliated Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210096, China
- Key Laboratory of Developmental Genes and Human Disease of Ministry of Education, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Ming Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China.
| | - Hong Chang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China.
| | - Xiong-Jian Luo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650223, China.
- Department of Neurology, Affiliated Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210096, China.
- Zhongda Hospital, School of Life Sciences and Technology, Advanced Institute for Life and Health, Southeast University, Nanjing, Jiangsu, 210096, China.
| |
Collapse
|
15
|
12 months is a pivotal age for olfactory perceptual learning and its underlying neuronal plasticity in aging mice. Neurobiol Aging 2022; 114:73-83. [DOI: 10.1016/j.neurobiolaging.2022.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 03/04/2022] [Accepted: 03/06/2022] [Indexed: 11/23/2022]
|
16
|
A Sexually Dimorphic Olfactory Neuron Mediates Fixed Action Transition during Courtship Ritual in Drosophila melanogaster. J Neurosci 2021; 41:9732-9741. [PMID: 34649953 DOI: 10.1523/jneurosci.1168-21.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 10/03/2021] [Accepted: 10/06/2021] [Indexed: 11/21/2022] Open
Abstract
Animals perform a series of actions in a fixed order during ritualistic innate behaviors. Although command neurons and sensory pathways responding to external stimuli that trigger these behaviors have been identified, how each action is induced in a fixed order in response to multimodal sensory stimuli remains unclear. Here, the sexually dimorphic lateral antennal lobe tract projection neuron 4 (lPN4) in male Drosophila melanogaster mediates the expression of a fixed behavioral action pattern at the beginning of the courtship ritual, in which a male taps a female body and then extends a wing unilaterally to produce a courtship song. We found that blocking the synaptic output of lPN4 caused an increase in the ratio of male flies that extended a wing unilaterally without tapping the female body, whereas excitation of lPN4 suppressed the transition from the tapping phase to the unilateral wing extension phase. Real-time calcium imaging showed that lPN4 is activated by a volatile pheromone, palmitoleic acid, whose responses were inhibited by simultaneous gustatory stimulation with female cuticular hydrocarbons, showing the existence of an "AND-gate" for multimodal sensory inputs during male courtship behaviors. These results suggest that the function of lPN4 is to suppress unilateral wing extension while responding to a female smell, which is released by appropriate contact chemosensory inputs received when tapping a female. As the female smell also promotes male courtship behaviors, the olfactory system is ready to simultaneously promote and suppress the progress of courtship actions while responding to a female smell.SIGNIFICANCE STATEMENT Although it has been 80 years since Konrad Lorenz and Niko Tinbergen introduced how multiple acts comprising separate innate behaviors are released in a fixed order according to external stimuli, the neural circuits responsible for such fixed action patterns remain largely unknown. The male courtship behavior of Drosophila melanogaster is a good model to investigate how such a fixed behavioral sequence is determined in the brain. Here, we show that lateral antennal lobe tract projection neuron 4 (lPN4) in D. melanogaster functions as an "AND-gate" for volatile and contact chemosensory inputs, mediating the expression of tapping behaviors before unilateral wing extension during male courtship rituals.
Collapse
|
17
|
Nakata H, Iseki S, Mizokami A. Three-dimensional reconstruction of testis cords/seminiferous tubules. Reprod Med Biol 2021; 20:402-409. [PMID: 34646067 PMCID: PMC8499590 DOI: 10.1002/rmb2.12413] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 09/02/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Due to the development of novel equipment for the acquisition of two-dimensional serial images and software capable of displaying three-dimensional (3D) images from serial images, the accurate 3D reconstruction of organs and tissues has become possible. METHODS Based on published studies, this review summarizes techniques for the 3D reconstruction of the testis cords/seminiferous tubules, with special reference to our method using serial paraffin sections and 3D visualization software. MAIN FINDINGS The testes of mice, rats, and hamsters of various ages were 3D reconstructed and species and age differences in the structures of the testis cords/seminiferous tubules were analyzed. Our method is advantageous because conventional paraffin-embedded normal and pathological specimens may be utilized for the 3D analysis without the need for complicated and expensive equipment. CONCLUSION By further decreasing the time and labor required for the procedure and adding information on molecular localization, the technique for 3D reconstruction will contribute to the elucidation of not only the structures, but also the functions of various organs, including the testis.
Collapse
Affiliation(s)
- Hiroki Nakata
- Department of Histology and Cell Biology Graduate School of Medical Sciences Kanazawa University Kanazawa Japan
| | - Shoichi Iseki
- Department of Clinical Engineering Faculty of Health Sciences Komatsu University Komatsu Japan
| | - Atsushi Mizokami
- Department of Integrative Cancer Therapy and Urology Kanazawa University Graduate School of Medical Science Kanazawa Japan
| |
Collapse
|
18
|
Das R, Bhattacharjee S, Letcher JM, Harris JM, Nanda S, Foldi I, Lottes EN, Bobo HM, Grantier BD, Mihály J, Ascoli GA, Cox DN. Formin 3 directs dendritic architecture via microtubule regulation and is required for somatosensory nociceptive behavior. Development 2021; 148:dev187609. [PMID: 34322714 PMCID: PMC8380456 DOI: 10.1242/dev.187609] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 07/12/2021] [Indexed: 01/26/2023]
Abstract
Dendrite shape impacts functional connectivity and is mediated by organization and dynamics of cytoskeletal fibers. Identifying the molecular factors that regulate dendritic cytoskeletal architecture is therefore important in understanding the mechanistic links between cytoskeletal organization and neuronal function. We identified Formin 3 (Form3) as an essential regulator of cytoskeletal architecture in nociceptive sensory neurons in Drosophila larvae. Time course analyses reveal that Form3 is cell-autonomously required to promote dendritic arbor complexity. We show that form3 is required for the maintenance of a population of stable dendritic microtubules (MTs), and mutants exhibit defects in the localization of dendritic mitochondria, satellite Golgi, and the TRPA channel Painless. Form3 directly interacts with MTs via FH1-FH2 domains. Mutations in human inverted formin 2 (INF2; ortholog of form3) have been causally linked to Charcot-Marie-Tooth (CMT) disease. CMT sensory neuropathies lead to impaired peripheral sensitivity. Defects in form3 function in nociceptive neurons result in severe impairment of noxious heat-evoked behaviors. Expression of the INF2 FH1-FH2 domains partially recovers form3 defects in MTs and nocifensive behavior, suggesting conserved functions, thereby providing putative mechanistic insights into potential etiologies of CMT sensory neuropathies.
Collapse
Affiliation(s)
- Ravi Das
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| | | | - Jamin M. Letcher
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| | - Jenna M. Harris
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| | - Sumit Nanda
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA 22030, USA
| | - Istvan Foldi
- Biological Research Centre, Hungarian Academy of Sciences, Institute of Genetics, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Erin N. Lottes
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| | - Hansley M. Bobo
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| | | | - József Mihály
- Biological Research Centre, Hungarian Academy of Sciences, Institute of Genetics, MTA-SZBK NAP B Axon Growth and Regeneration Group, Temesvári krt. 62, Szeged H-6726, Hungary
| | - Giorgio A. Ascoli
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA 22030, USA
| | - Daniel N. Cox
- Neuroscience Institute, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
19
|
Forest J, Chalençon L, Midroit M, Terrier C, Caillé I, Sacquet J, Benetollo C, Martin K, Richard M, Didier A, Mandairon N. Role of Adult-Born Versus Preexisting Neurons Born at P0 in Olfactory Perception in a Complex Olfactory Environment in Mice. Cereb Cortex 2021; 30:534-549. [PMID: 31216001 DOI: 10.1093/cercor/bhz105] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 03/26/2019] [Accepted: 04/21/2019] [Indexed: 12/11/2022] Open
Abstract
Olfactory perceptual learning is defined as an improvement in the discrimination of perceptually close odorants after passive exposure to these odorants. In mice, simple olfactory perceptual learning involving the discrimination of two odorants depends on an increased number of adult-born neurons in the olfactory bulb, which refines the bulbar output. However, the olfactory environment is complex, raising the question of the adjustment of the bulbar network to multiple discrimination challenges. Perceptual learning of 1 to 6 pairs of similar odorants led to discrimination of all learned odor pairs. Increasing complexity did not increase adult-born neuron survival but enhanced the number of adult-born neurons responding to learned odorants and their spine density. Moreover, only complex learning induced morphological changes in neurons of the granule cell layer born during the first day of life (P0). Selective optogenetic inactivation of either population confirmed functional involvement of adult-born neurons regardless of the enrichment complexity, while preexisting neurons were required for complex discrimination only.
Collapse
Affiliation(s)
- Jérémy Forest
- INSERM, U1028; CNRS, UMR5292; Lyon Neuroscience Research Center, Neuroplasticity and Neuropathology of Olfactory Perception Team, Lyon, F-69000, France.,Claude Bernard University Lyon1 and University of Lyon, Lyon F-69000, France
| | - Laura Chalençon
- INSERM, U1028; CNRS, UMR5292; Lyon Neuroscience Research Center, Neuroplasticity and Neuropathology of Olfactory Perception Team, Lyon, F-69000, France.,Claude Bernard University Lyon1 and University of Lyon, Lyon F-69000, France
| | - Maëllie Midroit
- INSERM, U1028; CNRS, UMR5292; Lyon Neuroscience Research Center, Neuroplasticity and Neuropathology of Olfactory Perception Team, Lyon, F-69000, France.,Claude Bernard University Lyon1 and University of Lyon, Lyon F-69000, France
| | - Claire Terrier
- INSERM, U1028; CNRS, UMR5292; Lyon Neuroscience Research Center, Neuroplasticity and Neuropathology of Olfactory Perception Team, Lyon, F-69000, France.,Claude Bernard University Lyon1 and University of Lyon, Lyon F-69000, France
| | - Isabelle Caillé
- Sorbonne Universités, Université Pierre et Marie Curie-Paris 06, Centre National de la Recherche Scientifique, UMR8246, INSERM U1130, Institut de Biologie Paris Seine, Neuroscience Paris Seine, and Sorbonne Paris Cité, Université Paris Diderot-Paris 7, Paris, France
| | - Joëlle Sacquet
- INSERM, U1028; CNRS, UMR5292; Lyon Neuroscience Research Center, Neuroplasticity and Neuropathology of Olfactory Perception Team, Lyon, F-69000, France.,Claude Bernard University Lyon1 and University of Lyon, Lyon F-69000, France
| | - Claire Benetollo
- INSERM, U1028, CNRS, UMR5292, Lyon Neuroscience Research Center, Neurogenetic and Optogenetic Platform, University Lyon 1 and University of Lyon, Lyon F-69000, France
| | - Killian Martin
- INSERM, U1028; CNRS, UMR5292; Lyon Neuroscience Research Center, Neuroplasticity and Neuropathology of Olfactory Perception Team, Lyon, F-69000, France.,Claude Bernard University Lyon1 and University of Lyon, Lyon F-69000, France
| | - Marion Richard
- INSERM, U1028; CNRS, UMR5292; Lyon Neuroscience Research Center, Neuroplasticity and Neuropathology of Olfactory Perception Team, Lyon, F-69000, France.,Claude Bernard University Lyon1 and University of Lyon, Lyon F-69000, France
| | - Anne Didier
- INSERM, U1028; CNRS, UMR5292; Lyon Neuroscience Research Center, Neuroplasticity and Neuropathology of Olfactory Perception Team, Lyon, F-69000, France.,Claude Bernard University Lyon1 and University of Lyon, Lyon F-69000, France
| | - Nathalie Mandairon
- INSERM, U1028; CNRS, UMR5292; Lyon Neuroscience Research Center, Neuroplasticity and Neuropathology of Olfactory Perception Team, Lyon, F-69000, France.,Claude Bernard University Lyon1 and University of Lyon, Lyon F-69000, France
| |
Collapse
|
20
|
Bingham CS, Parent M, McIntyre CC. Histology-driven model of the macaque motor hyperdirect pathway. Brain Struct Funct 2021; 226:2087-2097. [PMID: 34091730 DOI: 10.1007/s00429-021-02307-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 05/22/2021] [Indexed: 11/28/2022]
Abstract
Emerging appreciation for the hyperdirect pathway (HDP) as an important cortical glutamatergic input to the subthalamic nucleus (STN) has motivated a wide range of recent investigations on its role in motor control, as well as the mechanisms of subthalamic deep brain stimulation (DBS). However, the pathway anatomy and terminal arbor morphometry by which the HDP links cortical and subthalamic activity are incompletely understood. One critical hindrance to advancing understanding is the lack of anatomically detailed population models which can help explain how HDP pathway anatomy and neuronal biophysics give rise to spatiotemporal patterns of stimulus-response activity observed in vivo. Therefore, the goal of this study was to establish a population model of motor HDP axons through application of generative algorithms constrained by recent histology and imaging data. The products of this effort include a de novo macaque brain atlas, detailed statistical analysis of histological reconstructions of macaque motor HDP axons, and the generation of 10,000 morphometrically constrained synthetic motor HDP axons. The synthetic HDP axons exhibited a 3.8% mean error with respect to parametric distributions of the fiber target volume, total length, number of bifurcations, bifurcation angles, meander angles, and segment lengths measured in BDA-labeled HDP axon reconstructions. As such, this large population of synthetic motor HDP axons represents an anatomically based foundation for biophysical simulations that can be coupled to electrophysiological and/or behavioral measurements, with the goal of better understanding the role of the HDP in motor system activity.
Collapse
Affiliation(s)
- Clayton S Bingham
- Department of Biomedical Engineering, Case Western Reserve University, 2103 Cornell Road, Rm 6224, Cleveland, OH, 44106, USA
| | - Martin Parent
- CERVO Brain Research Center, Department of Psychiatry and Neuroscience, Faculty of Medicine, University of Laval, Quebec, Canada
| | - Cameron C McIntyre
- Department of Biomedical Engineering, Case Western Reserve University, 2103 Cornell Road, Rm 6224, Cleveland, OH, 44106, USA.
| |
Collapse
|
21
|
Ikezu S, Yeh H, Delpech JC, Woodbury ME, Van Enoo AA, Ruan Z, Sivakumaran S, You Y, Holland C, Guillamon-Vivancos T, Yoshii-Kitahara A, Botros MB, Madore C, Chao PH, Desani A, Manimaran S, Kalavai SV, Johnson WE, Butovsky O, Medalla M, Luebke JI, Ikezu T. Inhibition of colony stimulating factor 1 receptor corrects maternal inflammation-induced microglial and synaptic dysfunction and behavioral abnormalities. Mol Psychiatry 2021; 26:1808-1831. [PMID: 32071385 PMCID: PMC7431382 DOI: 10.1038/s41380-020-0671-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 01/21/2020] [Accepted: 01/29/2020] [Indexed: 12/23/2022]
Abstract
Maternal immune activation (MIA) disrupts the central innate immune system during a critical neurodevelopmental period. Microglia are primary innate immune cells in the brain although their direct influence on the MIA phenotype is largely unknown. Here we show that MIA alters microglial gene expression with upregulation of cellular protrusion/neuritogenic pathways, concurrently causing repetitive behavior, social deficits, and synaptic dysfunction to layer V intrinsically bursting pyramidal neurons in the prefrontal cortex of mice. MIA increases plastic dendritic spines of the intrinsically bursting neurons and their interaction with hyper-ramified microglia. Treating MIA offspring by colony stimulating factor 1 receptor inhibitors induces depletion and repopulation of microglia, and corrects protein expression of the newly identified MIA-associated neuritogenic molecules in microglia, which coalesces with correction of MIA-associated synaptic, neurophysiological, and behavioral abnormalities. Our study demonstrates that maternal immune insults perturb microglial phenotypes and influence neuronal functions throughout adulthood, and reveals a potent effect of colony stimulating factor 1 receptor inhibitors on the correction of MIA-associated microglial, synaptic, and neurobehavioral dysfunctions.
Collapse
Affiliation(s)
- Seiko Ikezu
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.
| | - Hana Yeh
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
- Graduate Program in Neuroscience, Boston University, Boston, MA, USA
| | - Jean-Christophe Delpech
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Maya E Woodbury
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
- Graduate Program in Neuroscience, Boston University, Boston, MA, USA
| | - Alicia A Van Enoo
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Zhi Ruan
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Sudhir Sivakumaran
- Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | - Yang You
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Carl Holland
- Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | | | - Asuka Yoshii-Kitahara
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Mina B Botros
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Charlotte Madore
- Ann Romney Center for Neurologic Diseases, Department of Neurology and Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pin-Hao Chao
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Ankita Desani
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Solaiappan Manimaran
- Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Srinidhi Venkatesan Kalavai
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - W Evan Johnson
- Computational Biomedicine, Boston University School of Medicine, Boston, MA, USA
| | - Oleg Butovsky
- Ann Romney Center for Neurologic Diseases, Department of Neurology and Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Maria Medalla
- Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | - Jennifer I Luebke
- Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, USA
| | - Tsuneya Ikezu
- Departments of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA.
- Center for Systems Neuroscience, Boston University, Boston, MA, USA.
- Department of Neurology and Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
22
|
A morphological analysis of activity-dependent myelination and myelin injury in transitional oligodendrocytes. Sci Rep 2021; 11:9588. [PMID: 33953273 PMCID: PMC8099889 DOI: 10.1038/s41598-021-88887-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 04/07/2021] [Indexed: 12/13/2022] Open
Abstract
Neuronal activity is established as a driver of oligodendrocyte (OL) differentiation and myelination. The concept of activity-dependent myelin plasticity, and its role in cognition and disease, is gaining support. Methods capable of resolving changes in the morphology of individual myelinating OL would advance our understanding of myelin plasticity and injury, thus we adapted a labelling approach involving Semliki Forest Virus (SFV) vectors to resolve and quantify the 3-D structure of OL processes and internodes in cerebellar slice cultures. We first demonstrate the utility of the approach by studying changes in OL morphology after complement-mediated injury. SFV vectors injected into cerebellar white matter labelled transitional OL (TOL), whose characteristic mixture of myelinating and non-myelinating processes exhibited significant degeneration after complement injury. The method was also capable of resolving finer changes in morphology related to neuronal activity. Prolonged suppression of neuronal activity, which reduced myelination, selectively decreased the length of putative internodes, and the proportion of process branches that supported them, while leaving other features of process morphology unaltered. Overall this work provides novel information on the morphology of TOL, and their response to conditions that alter circuit function or induce demyelination.
Collapse
|
23
|
Bączyńska E, Pels KK, Basu S, Włodarczyk J, Ruszczycki B. Quantification of Dendritic Spines Remodeling under Physiological Stimuli and in Pathological Conditions. Int J Mol Sci 2021; 22:4053. [PMID: 33919977 PMCID: PMC8070910 DOI: 10.3390/ijms22084053] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 12/14/2022] Open
Abstract
Numerous brain diseases are associated with abnormalities in morphology and density of dendritic spines, small membranous protrusions whose structural geometry correlates with the strength of synaptic connections. Thus, the quantitative analysis of dendritic spines remodeling in microscopic images is one of the key elements towards understanding mechanisms of structural neuronal plasticity and bases of brain pathology. In the following article, we review experimental approaches designed to assess quantitative features of dendritic spines under physiological stimuli and in pathological conditions. We compare various methodological pipelines of biological models, sample preparation, data analysis, image acquisition, sample size, and statistical analysis. The methodology and results of relevant experiments are systematically summarized in a tabular form. In particular, we focus on quantitative data regarding the number of animals, cells, dendritic spines, types of studied parameters, size of observed changes, and their statistical significance.
Collapse
Affiliation(s)
- Ewa Bączyńska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (E.B.); (K.K.P.); (J.W.)
| | - Katarzyna Karolina Pels
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (E.B.); (K.K.P.); (J.W.)
| | - Subhadip Basu
- Department of Computer Science and Engineering, Jadvapur University, Kolkata 700032, India;
| | - Jakub Włodarczyk
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (E.B.); (K.K.P.); (J.W.)
| | - Błażej Ruszczycki
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (E.B.); (K.K.P.); (J.W.)
| |
Collapse
|
24
|
Nowzari F, Wang H, Khoradmehr A, Baghban M, Baghban N, Arandian A, Muhaddesi M, Nabipour I, Zibaii MI, Najarasl M, Taheri P, Latifi H, Tamadon A. Three-Dimensional Imaging in Stem Cell-Based Researches. Front Vet Sci 2021; 8:657525. [PMID: 33937378 PMCID: PMC8079735 DOI: 10.3389/fvets.2021.657525] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 03/19/2021] [Indexed: 12/14/2022] Open
Abstract
Stem cells have an important role in regenerative therapies, developmental biology studies and drug screening. Basic and translational research in stem cell technology needs more detailed imaging techniques. The possibility of cell-based therapeutic strategies has been validated in the stem cell field over recent years, a more detailed characterization of the properties of stem cells is needed for connectomics of large assemblies and structural analyses of these cells. The aim of stem cell imaging is the characterization of differentiation state, cellular function, purity and cell location. Recent progress in stem cell imaging field has included ultrasound-based technique to study living stem cells and florescence microscopy-based technique to investigate stem cell three-dimensional (3D) structures. Here, we summarized the fundamental characteristics of stem cells via 3D imaging methods and also discussed the emerging literatures on 3D imaging in stem cell research and the applications of both classical 2D imaging techniques and 3D methods on stem cells biology.
Collapse
Affiliation(s)
- Fariborz Nowzari
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Huimei Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Arezoo Khoradmehr
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mandana Baghban
- Department of Obstetrics and Gynecology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Neda Baghban
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Alireza Arandian
- Laser and Plasma Research Institute, Shahid Beheshti University, Tehran, Iran
| | - Mahdi Muhaddesi
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Iraj Nabipour
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mohammad I. Zibaii
- Laser and Plasma Research Institute, Shahid Beheshti University, Tehran, Iran
| | - Mostafa Najarasl
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Payam Taheri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Hamid Latifi
- Laser and Plasma Research Institute, Shahid Beheshti University, Tehran, Iran
- Department of Physics, Shahid Beheshti University, Tehran, Iran
| | - Amin Tamadon
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| |
Collapse
|
25
|
Betaine ameliorates schizophrenic traits by functionally compensating for KIF3-based CRMP2 transport. Cell Rep 2021; 35:108971. [PMID: 33852848 DOI: 10.1016/j.celrep.2021.108971] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 12/22/2020] [Accepted: 03/19/2021] [Indexed: 12/13/2022] Open
Abstract
In schizophrenia (SCZ), neurons in the brain tend to undergo gross morphological changes, but the related molecular mechanism remains largely elusive. Using Kif3b+/- mice as a model with SCZ-like behaviors, we found that a high-betaine diet can significantly alleviate schizophrenic traits related to neuronal morphogenesis and behaviors. According to a deficiency in the transport of collapsin response mediator protein 2 (CRMP2) by the KIF3 motor, we identified a significant reduction in lamellipodial dynamics in developing Kif3b+/- neurons as a cause of neurite hyperbranching. Betaine administration significantly decreases CRMP2 carbonylation, which enhances the F-actin bundling needed for proper lamellipodial dynamics and microtubule exclusion and may thus functionally compensate for KIF3 deficiency. Because the KIF3 expression levels tend to be downregulated in the human prefrontal cortex of the postmortem brains of SCZ patients, this mechanism may partly participate in human SCZ pathogenesis, which we hypothesize could be alleviated by betaine administration.
Collapse
|
26
|
Salesse C, Charest J, Doucet-Beaupré H, Castonguay AM, Labrecque S, De Koninck P, Lévesque M. Opposite Control of Excitatory and Inhibitory Synapse Formation by Slitrk2 and Slitrk5 on Dopamine Neurons Modulates Hyperactivity Behavior. Cell Rep 2021; 30:2374-2386.e5. [PMID: 32075770 DOI: 10.1016/j.celrep.2020.01.084] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 12/03/2019] [Accepted: 01/24/2020] [Indexed: 11/26/2022] Open
Abstract
The neurodevelopmental origin of hyperactivity disorder has been suggested to involve the dopaminergic system, but the underlying mechanisms are still unknown. Here, transcription factors Lmx1a and Lmx1b are shown to be essential for midbrain dopaminergic (mDA) neuron excitatory synaptic inputs and dendritic development. Strikingly, conditional knockout (cKO) of Lmx1a/b in postmitotic mDA neurons results in marked hyperactivity. In seeking Lmx1a/b target genes, we identify positively regulated Slitrk2 and negatively regulated Slitrk5. These two synaptic adhesion proteins promote excitatory and inhibitory synapses on mDA neurons, respectively. Knocking down Slitrk2 reproduces some of the Lmx1a/b cKO cellular and behavioral phenotypes, whereas Slitrk5 knockdown has opposite effects. The hyperactivity caused by this imbalance in excitatory/inhibitory synaptic inputs on dopamine neurons is reproduced by chronically inhibiting the ventral tegmental area during development using pharmacogenetics. Our study shows that alterations in developing dopaminergic circuits strongly impact locomotor activity, shedding light on mechanisms causing hyperactivity behaviors.
Collapse
Affiliation(s)
- Charleen Salesse
- CERVO Brain Research Centre, 2601 de la Canardière, Québec, QC G1J 2G3, Canada
| | - Julien Charest
- CERVO Brain Research Centre, 2601 de la Canardière, Québec, QC G1J 2G3, Canada
| | | | | | - Simon Labrecque
- CERVO Brain Research Centre, 2601 de la Canardière, Québec, QC G1J 2G3, Canada
| | - Paul De Koninck
- CERVO Brain Research Centre, 2601 de la Canardière, Québec, QC G1J 2G3, Canada; Department of Biochemistry, Microbiology and Bioinformatics, Faculty of Science and Engineering, Université Laval, Québec, QC G1V 0A6, Canada
| | - Martin Lévesque
- CERVO Brain Research Centre, 2601 de la Canardière, Québec, QC G1J 2G3, Canada; Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada.
| |
Collapse
|
27
|
Sánchez-González A, Thougaard E, Tapias-Espinosa C, Cañete T, Sampedro-Viana D, Saunders JM, Toneatti R, Tobeña A, Gónzalez-Maeso J, Aznar S, Fernández-Teruel A. Increased thin-spine density in frontal cortex pyramidal neurons in a genetic rat model of schizophrenia-relevant features. Eur Neuropsychopharmacol 2021; 44:79-91. [PMID: 33485732 PMCID: PMC7902438 DOI: 10.1016/j.euroneuro.2021.01.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 12/09/2020] [Accepted: 01/10/2021] [Indexed: 12/13/2022]
Abstract
The cellular mechanisms altered during brain wiring leading to cognitive disturbances in neurodevelopmental disorders remain unknown. We have previously reported altered cortical expression of neurodevelopmentally regulated synaptic markers in a genetic animal model of schizophrenia-relevant behavioral features, the Roman-High Avoidance rat strain (RHA-I). To further explore this phenotype, we looked at dendritic spines in cortical pyramidal neurons, as changes in spine density and morphology are one of the main processes taking place during adolescence. An HSV-viral vector carrying green fluorescent protein (GFP) was injected into the frontal cortex (FC) of a group of 11 RHA-I and 12 Roman-Low Avoidance (RLA-I) male rats. GFP labeled dendrites from pyramidal cells were 3D reconstructed and number and types of spines quantified. We observed an increased spine density in the RHA-I, corresponding to a larger fraction of immature thin spines, with no differences in stubby and mushroom spines. Glia cells, parvalbumin (PV) and somatostatin (SST) interneurons and surrounding perineuronal net (PNN) density are known to participate in FC and pyramidal neuron dendritic spine maturation. We determined by stereological-based quantification a significantly higher number of GFAP-positive astrocytes in the FC of the RHA-I strain, with no difference in microglia (Iba1-positive cells). The number of inhibitory PV, SST interneurons or PNN density, on the contrary, was unchanged. Results support our belief that the RHA-I strain presents a more immature FC, with some structural features like those observed during adolescence, adding construct validity to this strain as a genetic behavioral model of neurodevelopmental disorders.
Collapse
Affiliation(s)
- A Sánchez-González
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, School of Medicine, Universidad Autónoma de Barcelona, 08193 Bellaterra, Barcelona, Spain; Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - E Thougaard
- Research Laboratory for Stereology and Neuroscience, Bispebjerg Copenhagen University Hospital, 2400 Copenhagen, Denmark
| | - C Tapias-Espinosa
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, School of Medicine, Universidad Autónoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - T Cañete
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, School of Medicine, Universidad Autónoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - D Sampedro-Viana
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, School of Medicine, Universidad Autónoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - J M Saunders
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - R Toneatti
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - A Tobeña
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, School of Medicine, Universidad Autónoma de Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - J Gónzalez-Maeso
- Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - S Aznar
- Research Laboratory for Stereology and Neuroscience, Bispebjerg Copenhagen University Hospital, 2400 Copenhagen, Denmark; Copenhagen Center for Translational Research, Bispebjerg Copenhagen University Hospital, Copenhagen, Denmark.
| | - A Fernández-Teruel
- Medical Psychology Unit, Department of Psychiatry and Forensic Medicine, Institute of Neurosciences, School of Medicine, Universidad Autónoma de Barcelona, 08193 Bellaterra, Barcelona, Spain.
| |
Collapse
|
28
|
Jafari M, Schumacher AM, Snaidero N, Ullrich Gavilanes EM, Neziraj T, Kocsis-Jutka V, Engels D, Jürgens T, Wagner I, Weidinger JDF, Schmidt SS, Beltrán E, Hagan N, Woodworth L, Ofengeim D, Gans J, Wolf F, Kreutzfeldt M, Portugues R, Merkler D, Misgeld T, Kerschensteiner M. Phagocyte-mediated synapse removal in cortical neuroinflammation is promoted by local calcium accumulation. Nat Neurosci 2021; 24:355-367. [PMID: 33495636 DOI: 10.1038/s41593-020-00780-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/16/2020] [Indexed: 01/30/2023]
Abstract
Cortical pathology contributes to chronic cognitive impairment of patients suffering from the neuroinflammatory disease multiple sclerosis (MS). How such gray matter inflammation affects neuronal structure and function is not well understood. In the present study, we use functional and structural in vivo imaging in a mouse model of cortical MS to demonstrate that bouts of cortical inflammation disrupt cortical circuit activity coincident with a widespread, but transient, loss of dendritic spines. Spines destined for removal show local calcium accumulations and are subsequently removed by invading macrophages or activated microglia. Targeting phagocyte activation with a new antagonist of the colony-stimulating factor 1 receptor prevents cortical synapse loss. Overall, our study identifies synapse loss as a key pathological feature of inflammatory gray matter lesions that is amenable to immunomodulatory therapy.
Collapse
Affiliation(s)
- Mehrnoosh Jafari
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany.,Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University Munich, Munich, Germany
| | - Adrian-Minh Schumacher
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany.,Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University Munich, Munich, Germany
| | - Nicolas Snaidero
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany.,Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany.,German Center for Neurodegenerative Diseases, Munich, Germany
| | - Emily M Ullrich Gavilanes
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany.,Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University Munich, Munich, Germany
| | - Tradite Neziraj
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany.,Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University Munich, Munich, Germany
| | - Virág Kocsis-Jutka
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany.,Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University Munich, Munich, Germany
| | - Daniel Engels
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany.,Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University Munich, Munich, Germany
| | - Tanja Jürgens
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Ingrid Wagner
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
| | - Juan Daniel Flórez Weidinger
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany.,Bernstein Center for Computational Neuroscience, University of Göttingen, Göttingen, Germany.,Max Planck Institute for Experimental Medicine, Göttingen, Germany.,Center for Biostructural Imaging of Neurodegeneration, Göttingen, Germany
| | - Stephanie S Schmidt
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Eduardo Beltrán
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany.,Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University Munich, Munich, Germany
| | - Nellwyn Hagan
- Rare and Neurological Disease Research, Sanofi, Framingham, MA, USA
| | - Lisa Woodworth
- Rare and Neurological Disease Research, Sanofi, Framingham, MA, USA
| | - Dimitry Ofengeim
- Rare and Neurological Disease Research, Sanofi, Framingham, MA, USA
| | - Joseph Gans
- Translational Sciences Genomics, Sanofi, Framingham, MA, USA
| | - Fred Wolf
- Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany.,Bernstein Center for Computational Neuroscience, University of Göttingen, Göttingen, Germany.,Max Planck Institute for Experimental Medicine, Göttingen, Germany.,Center for Biostructural Imaging of Neurodegeneration, Göttingen, Germany.,Campus Institute for Dynamics of Biological Networks, University of Göttingen, Göttingen, Germany
| | - Mario Kreutzfeldt
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland.,Division of Clinical Pathology, Geneva University Hospital, Geneva, Switzerland
| | - Ruben Portugues
- Sensorimotor Control, Max Planck Institute of Neurobiology, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Doron Merkler
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland. .,Division of Clinical Pathology, Geneva University Hospital, Geneva, Switzerland.
| | - Thomas Misgeld
- Institute of Neuronal Cell Biology, Technical University of Munich, Munich, Germany. .,German Center for Neurodegenerative Diseases, Munich, Germany. .,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| | - Martin Kerschensteiner
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany. .,Biomedical Center, Faculty of Medicine, Ludwig-Maximilians University Munich, Munich, Germany. .,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
29
|
How neurons exploit fractal geometry to optimize their network connectivity. Sci Rep 2021; 11:2332. [PMID: 33504818 PMCID: PMC7840685 DOI: 10.1038/s41598-021-81421-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/30/2020] [Indexed: 11/13/2022] Open
Abstract
We investigate the degree to which neurons are fractal, the origin of this fractality, and its impact on functionality. By analyzing three-dimensional images of rat neurons, we show the way their dendrites fork and weave through space is unexpectedly important for generating fractal-like behavior well-described by an ‘effective’ fractal dimension D. This discovery motivated us to create distorted neuron models by modifying the dendritic patterns, so generating neurons across wide ranges of D extending beyond their natural values. By charting the D-dependent variations in inter-neuron connectivity along with the associated costs, we propose that their D values reflect a network cooperation that optimizes these constraints. We discuss the implications for healthy and pathological neurons, and for connecting neurons to medical implants. Our automated approach also facilitates insights relating form and function, applicable to individual neurons and their networks, providing a crucial tool for addressing massive data collection projects (e.g. connectomes).
Collapse
|
30
|
Disruption of NEUROD2 causes a neurodevelopmental syndrome with autistic features via cell-autonomous defects in forebrain glutamatergic neurons. Mol Psychiatry 2021; 26:6125-6148. [PMID: 34188164 PMCID: PMC8760061 DOI: 10.1038/s41380-021-01179-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 05/17/2021] [Accepted: 05/26/2021] [Indexed: 02/06/2023]
Abstract
While the transcription factor NEUROD2 has recently been associated with epilepsy, its precise role during nervous system development remains unclear. Using a multi-scale approach, we set out to understand how Neurod2 deletion affects the development of the cerebral cortex in mice. In Neurod2 KO embryos, cortical projection neurons over-migrated, thereby altering the final size and position of layers. In juvenile and adults, spine density and turnover were dysregulated in apical but not basal compartments in layer 5 neurons. Patch-clamp recordings in layer 5 neurons of juvenile mice revealed increased intrinsic excitability. Bulk RNA sequencing showed dysregulated expression of many genes associated with neuronal excitability and synaptic function, whose human orthologs were strongly associated with autism spectrum disorders (ASD). At the behavior level, Neurod2 KO mice displayed social interaction deficits, stereotypies, hyperactivity, and occasionally spontaneous seizures. Mice heterozygous for Neurod2 had similar defects, indicating that Neurod2 is haploinsufficient. Finally, specific deletion of Neurod2 in forebrain excitatory neurons recapitulated cellular and behavioral phenotypes found in constitutive KO mice, revealing the region-specific contribution of dysfunctional Neurod2 in symptoms. Informed by these neurobehavioral features in mouse mutants, we identified eleven patients from eight families with a neurodevelopmental disorder including intellectual disability and ASD associated with NEUROD2 pathogenic mutations. Our findings demonstrate crucial roles for Neurod2 in neocortical development, whose alterations can cause neurodevelopmental disorders including intellectual disability and ASD.
Collapse
|
31
|
Kontou G, Josephine Ng SF, Cardarelli RA, Howden JH, Choi C, Ren Q, Rodriguez Santos MA, Bope CE, Dengler JS, Kelley MR, Davies PA, Kittler JT, Brandon NJ, Moss SJ, Smalley JL. KCC2 is required for the survival of mature neurons but not for their development. J Biol Chem 2021; 296:100364. [PMID: 33539918 PMCID: PMC7949141 DOI: 10.1016/j.jbc.2021.100364] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/24/2021] [Accepted: 01/29/2021] [Indexed: 12/11/2022] Open
Abstract
The K+/Cl- cotransporter KCC2 (SLC12A5) allows mature neurons in the CNS to maintain low intracellular Cl- levels that are critical in mediating fast hyperpolarizing synaptic inhibition via type A γ-aminobutyric acid receptors (GABAARs). In accordance with this, compromised KCC2 activity results in seizures, but whether such deficits directly contribute to the subsequent changes in neuronal structure and viability that lead to epileptogenesis remains to be assessed. Canonical hyperpolarizing GABAAR currents develop postnatally, which reflect a progressive increase in KCC2 expression levels and activity. To investigate the role that KCC2 plays in regulating neuronal viability and architecture, we have conditionally ablated KCC2 expression in developing and mature neurons. Decreasing KCC2 expression in mature neurons resulted in the rapid activation of the extrinsic apoptotic pathway. Intriguingly, direct pharmacological inhibition of KCC2 in mature neurons was sufficient to rapidly induce apoptosis, an effect that was not abrogated via blockade of neuronal depolarization using tetrodotoxin (TTX). In contrast, ablating KCC2 expression in immature neurons had no discernable effects on their subsequent development, arborization, or dendritic structure. However, removing KCC2 in immature neurons was sufficient to ablate the subsequent postnatal development of hyperpolarizing GABAAR currents. Collectively, our results demonstrate that KCC2 plays a critical role in neuronal survival by limiting apoptosis, and mature neurons are highly sensitive to the loss of KCC2 function. In contrast, KCC2 appears to play a minimal role in mediating neuronal development or architecture.
Collapse
Affiliation(s)
- Georgina Kontou
- AstraZeneca-Tufts Laboratory of Basic and Translational Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA; Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Shu Fun Josephine Ng
- AstraZeneca-Tufts Laboratory of Basic and Translational Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA; Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Ross A Cardarelli
- AstraZeneca-Tufts Laboratory of Basic and Translational Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA; Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Jack H Howden
- Department of Neuroscience, Physiology, and Pharmacology, University College London, London, UK
| | - Catherine Choi
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Qiu Ren
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA
| | | | - Christopher E Bope
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Jake S Dengler
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Matt R Kelley
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Paul A Davies
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Josef T Kittler
- Department of Neuroscience, Physiology, and Pharmacology, University College London, London, UK
| | - Nicholas J Brandon
- AstraZeneca-Tufts Laboratory of Basic and Translational Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA; Neuroscience, IMED Biotech Unit, AstraZeneca, Boston, Massachusetts, USA
| | - Stephen J Moss
- AstraZeneca-Tufts Laboratory of Basic and Translational Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA; Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA; Department of Neuroscience, Physiology, and Pharmacology, University College London, London, UK.
| | - Joshua L Smalley
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
32
|
Kirch C, Gollo LL. Spatially resolved dendritic integration: towards a functional classification of neurons. PeerJ 2020; 8:e10250. [PMID: 33282551 PMCID: PMC7694565 DOI: 10.7717/peerj.10250] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 10/06/2020] [Indexed: 01/19/2023] Open
Abstract
The vast tree-like dendritic structure of neurons allows them to receive and integrate input from many neurons. A wide variety of neuronal morphologies exist, however, their role in dendritic integration, and how it shapes the response of the neuron, is not yet fully understood. Here, we study the evolution and interactions of dendritic spikes in excitable neurons with complex real branch structures. We focus on dozens of digitally reconstructed illustrative neurons from the online repository NeuroMorpho.org, which contains over 130,000 neurons. Yet, our methods can be promptly extended to any other neuron. This approach allows us to estimate and map specific and heterogeneous patterns of activity observed across extensive dendritic trees with thousands of compartments. We propose a classification of neurons based on the location of the soma (centrality) and the number of branches connected to the soma. These are key topological factors in determining the neuron's energy consumption, firing rate, and the dynamic range, which quantifies the range in synaptic input rate that can be reliably encoded by the neuron's firing rate. Moreover, we find that bifurcations, the structural building blocks of complex dendrites, play a major role in increasing the dynamic range of neurons. Our results provide a better understanding of the effects of neuronal morphology in the diversity of neuronal dynamics and function.
Collapse
Affiliation(s)
- Christoph Kirch
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Queensland University of Technology, Brisbane, QLD, Australia
| | - Leonardo L. Gollo
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Queensland University of Technology, Brisbane, QLD, Australia
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
33
|
Anastasiades PG, Boada C, Carter AG. Cell-Type-Specific D1 Dopamine Receptor Modulation of Projection Neurons and Interneurons in the Prefrontal Cortex. Cereb Cortex 2020; 29:3224-3242. [PMID: 30566584 DOI: 10.1093/cercor/bhy299] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 11/01/2018] [Accepted: 11/07/2018] [Indexed: 11/14/2022] Open
Abstract
Dopamine modulation in the prefrontal cortex (PFC) mediates diverse effects on neuronal physiology and function, but the expression of dopamine receptors at subpopulations of projection neurons and interneurons remains unresolved. Here, we examine D1 receptor expression and modulation at specific cell types and layers in the mouse prelimbic PFC. We first show that D1 receptors are enriched in pyramidal cells in both layers 5 and 6, and that these cells project to intratelencephalic targets including contralateral cortex, striatum, and claustrum rather than to extratelencephalic structures. We then find that D1 receptors are also present in interneurons and enriched in superficial layer VIP-positive (VIP+) interneurons that coexpresses calretinin but absent from parvalbumin-positive (PV+) and somatostatin-positive (SOM+) interneurons. Finally, we determine that D1 receptors strongly and selectively enhance action potential firing in only a subset of these corticocortical neurons and VIP+ interneurons. Our findings define several novel subpopulations of D1+ neurons, highlighting how modulation via D1 receptors can influence both excitatory and disinhibitory microcircuits in the PFC.
Collapse
Affiliation(s)
- Paul G Anastasiades
- Center for Neural Science, New York University, 4 Washington Place, New York, NY, USA
| | - Christina Boada
- Center for Neural Science, New York University, 4 Washington Place, New York, NY, USA
| | - Adam G Carter
- Center for Neural Science, New York University, 4 Washington Place, New York, NY, USA
| |
Collapse
|
34
|
Pilcher W, Yang X, Zhurikhina A, Chernaya O, Xu Y, Qiu P, Tsygankov D. Shape-to-graph mapping method for efficient characterization and classification of complex geometries in biological images. PLoS Comput Biol 2020; 16:e1007758. [PMID: 32881897 PMCID: PMC7494120 DOI: 10.1371/journal.pcbi.1007758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 09/16/2020] [Accepted: 07/16/2020] [Indexed: 11/18/2022] Open
Abstract
With the ever-increasing quality and quantity of imaging data in biomedical research comes the demand for computational methodologies that enable efficient and reliable automated extraction of the quantitative information contained within these images. One of the challenges in providing such methodology is the need for tailoring algorithms to the specifics of the data, limiting their areas of application. Here we present a broadly applicable approach to quantification and classification of complex shapes and patterns in biological or other multi-component formations. This approach integrates the mapping of all shape boundaries within an image onto a global information-rich graph and machine learning on the multidimensional measures of the graph. We demonstrated the power of this method by (1) extracting subtle structural differences from visually indistinguishable images in our phenotype rescue experiments using the endothelial tube formations assay, (2) training the algorithm to identify biophysical parameters underlying the formation of different multicellular networks in our simulation model of collective cell behavior, and (3) analyzing the response of U2OS cell cultures to a broad array of small molecule perturbations. In this paper, we present a methodology that is based on mapping an arbitrary set of outlines onto a complete, strictly defined structure, in which every point representing the shape becomes a terminal point of a global graph. Because this mapping preserves the whole complexity of the shape, it allows for extracting the full scope of geometric features of any scale. Importantly, an extensive set of graph-based metrics in each image makes integration with machine learning routines highly efficient even for a small data sets and provide an opportunity to backtrack the subtle morphological features responsible for the automated distinction into image classes. The resulting tool provides efficient, versatile, and robust quantification of complex shapes and patterns in experimental images.
Collapse
Affiliation(s)
- William Pilcher
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Xingyu Yang
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Anastasia Zhurikhina
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Olga Chernaya
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Yinghan Xu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Peng Qiu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Denis Tsygankov
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
35
|
Werginz P, Raghuram V, Fried SI. Tailoring of the axon initial segment shapes the conversion of synaptic inputs into spiking output in OFF-α T retinal ganglion cells. SCIENCE ADVANCES 2020; 6:6/37/eabb6642. [PMID: 32917708 PMCID: PMC7486099 DOI: 10.1126/sciadv.abb6642] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/29/2020] [Indexed: 06/11/2023]
Abstract
Recently, mouse OFF-α transient (OFF-α T) retinal ganglion cells (RGCs) were shown to display a gradient of light responses as a function of position along the dorsal-ventral axis; response differences were correlated to differences in the level of excitatory presynaptic input. Here, we show that postsynaptic differences between cells also make a strong contribution to response differences. Cells in the dorsal retina had longer axon initial segments (AISs)-the greater number of Nav1.6 channels in longer AISs directly mediates higher rates of spiking and helps avoid depolarization block that terminates spiking in ventral cells with shorter AISs. The pre- and postsynaptic specializations that shape the output of OFF-α T RGCs interact in different ways: In dorsal cells, strong inputs and the long AISs are both necessary to generate their strong, sustained spiking outputs, while in ventral cells, weak inputs or the short AISs are both sufficient to limit the spiking signal.
Collapse
Affiliation(s)
- Paul Werginz
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Institute for Analysis and Scientific Computing, Vienna University of Technology, 1040 Vienna, Austria
| | - Vineeth Raghuram
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Boston VA Healthcare System, Rehabilitation, Research and Development, Boston, MA 02130, USA
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Shelley I Fried
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
- Boston VA Healthcare System, Rehabilitation, Research and Development, Boston, MA 02130, USA
| |
Collapse
|
36
|
Werginz P, Raghuram V, Fried SI. The relationship between morphological properties and thresholds to extracellular electric stimulation in α RGCs. J Neural Eng 2020; 17:045015. [PMID: 32736374 DOI: 10.1088/1741-2552/abab47] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Retinal prostheses strive to restore vison to patients that are blind from retinal degeneration by electrically stimulating surviving retinal ganglion cells (RGCs). The quality of elicited percepts remains limited however and it is desirable to develop improved stimulation strategies. Here, we examine how the anatomical and biophysical properties of RGCs influence activation thresholds, including the effects of variations found naturally. APPROACH Detailed reconstructions were made of a large number of mouse α RGCs and were used to create an array of model cells; the models were used to study the effects of individual anatomical features on activation threshold to electric stimulation. Stimulation was delivered epiretinally from a point-source or disk electrode and consisted of monophasic or biphasic rectangular pulses. MAIN RESULTS Modeling results show that the region of minimum threshold always is within the axon initial segment (AIS). The properties of this region as well as the absolute value of the minimum threshold are dependent on the length of the AIS as well as on the relative composition of sodium channels within the AIS. Other morphological features, including cell size, dendritic field size and the distance between the AIS and the soma had only a minimal influence on thresholds. Introducing even a small number of low-threshold Nav1.6 channels into the AIS was sufficient to lower minimum thresholds substantially although further increases in Nav1.6 had diminishing effects. The distance between the AIS and the electrode affects threshold levels while alignment of the electrode with the axon or dendritic parts of the RGC can result in lower thresholds, even if the distance to the cell remains the same. SIGNIFICANCE Intrinsic morphological features can influence activation thresholds with the AIS having the strongest influence. However, the combined influence remains limited and may not be large enough to allow for selective activation between different RGC types.
Collapse
Affiliation(s)
- Paul Werginz
- Institute for Analysis and Scientific Computing, Vienna University of Technology, Vienna, Austria. Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America. Author to whom any correspondence should be addressed
| | | | | |
Collapse
|
37
|
Zhu C, Mahmood Z, Zhang W, Akram MW, Ainur D, Ma H. In situ investigation of acute exposure of graphene oxide on activated sludge: Biofilm characteristics, microbial activity and cytotoxicity. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 199:110639. [PMID: 32408033 DOI: 10.1016/j.ecoenv.2020.110639] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/26/2020] [Accepted: 04/10/2020] [Indexed: 06/11/2023]
Abstract
Graphene Oxide (GO) has wide applications in many fields which has caused a large expected quantity of the graphene-based wastes. It is necessary to understand the toxic effects of the GO on the activated sludge (AS) considering its inevitable discharge to the wastewater treatment plants as the ultimate repositories for these wastes. In this study, the acute exposures of the multilayer Nano-graphene oxide (MNGO) at different dosages were conducted in order to investigate its integrated effects on the formation of the biofilm, mature biofilm and the microbial activity of the activated sludge. Raman spectroscopy and laser scanning confocal microscopy (LSCM) were adopted for the in-situ characterization of the biofilm with the exposure of the MNGO. The results showed that the activated sludge was tolerable to the acute exposure of the less than 100 mg/L of the MNGO, especially for the mature biofilm, and only a subtle decrease was found in the size and thickness during the formation of the biofilm, while the amount of 300 mg/L of the MNGO caused the sever deterioration on the activated sludge system. The microbial metabolic activity, viability, and the biological removal of the nutrients were significantly affected with the more than 100 mg/L of the MNGO. It was also demonstrated by the microbial cytotoxicity tests that the increase in the exposure of the MNGO was related to the increase in the reactive oxygen species (ROS) and the damaging degree of the cell membrane.
Collapse
Affiliation(s)
- Chao Zhu
- School of Environmental Science & Engineering. Shaanxi University of Science and Technology, Xi'an, China.
| | - Zarak Mahmood
- School of Environmental Science & Engineering. Shaanxi University of Science and Technology, Xi'an, China
| | - Wenting Zhang
- School of Environmental Science & Engineering. Shaanxi University of Science and Technology, Xi'an, China
| | - M Waqar Akram
- Department of Precision Machinery and Instrumentation. University of Science and Technology of China, Hefei, China
| | - Dyussenova Ainur
- School of Environmental Science & Engineering. Shaanxi University of Science and Technology, Xi'an, China
| | - Hongrui Ma
- School of Environmental Science & Engineering. Shaanxi University of Science and Technology, Xi'an, China
| |
Collapse
|
38
|
Foggetti A, Baccini G, Arnold P, Schiffelholz T, Wulff P. Spiny and Non-spiny Parvalbumin-Positive Hippocampal Interneurons Show Different Plastic Properties. Cell Rep 2020; 27:3725-3732.e5. [PMID: 31242406 PMCID: PMC6613043 DOI: 10.1016/j.celrep.2019.05.098] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 04/06/2019] [Accepted: 05/22/2019] [Indexed: 01/14/2023] Open
Abstract
Dendritic spines control synaptic transmission and plasticity by augmenting post-synaptic potentials and providing biochemical compartmentalization. In principal cells, spines cover the dendritic tree at high densities, receive the overwhelming majority of excitatory inputs, and undergo experience-dependent structural re-organization. Although GABAergic interneurons have long been considered to be devoid of spines, a number of studies have reported the sparse existence of spines in interneurons. However, little is known about their organization or function at the cellular and network level. Here, we show that a subset of hippocampal parvalbumin-positive interneurons forms numerous dendritic spines with highly variable densities and input-selective organization. These spines form in areas with reduced perineuronal net sheathing, predispose for plastic changes in protein expression, and show input-specific re-organization after behavioral experience.
Collapse
Affiliation(s)
- Angelica Foggetti
- Institute of Physiology, Christian-Albrechts-University Kiel, 24098 Kiel, Germany.
| | - Gilda Baccini
- Institute of Physiology, Christian-Albrechts-University Kiel, 24098 Kiel, Germany
| | - Philipp Arnold
- Anatomical Institute, Christian-Albrechts-University Kiel, 24118 Kiel, Germany
| | - Thomas Schiffelholz
- Department of Psychiatry and Psychotherapy, Christian-Albrechts-University Kiel, 24105 Kiel, Germany
| | - Peer Wulff
- Institute of Physiology, Christian-Albrechts-University Kiel, 24098 Kiel, Germany.
| |
Collapse
|
39
|
Bertan F, Wischhof L, Sosulina L, Mittag M, Dalügge D, Fornarelli A, Gardoni F, Marcello E, Di Luca M, Fuhrmann M, Remy S, Bano D, Nicotera P. Loss of Ryanodine Receptor 2 impairs neuronal activity-dependent remodeling of dendritic spines and triggers compensatory neuronal hyperexcitability. Cell Death Differ 2020; 27:3354-3373. [PMID: 32641776 PMCID: PMC7853040 DOI: 10.1038/s41418-020-0584-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/15/2020] [Accepted: 06/17/2020] [Indexed: 12/17/2022] Open
Abstract
Dendritic spines are postsynaptic domains that shape structural and functional properties of neurons. Upon neuronal activity, Ca2+ transients trigger signaling cascades that determine the plastic remodeling of dendritic spines, which modulate learning and memory. Here, we study in mice the role of the intracellular Ca2+ channel Ryanodine Receptor 2 (RyR2) in synaptic plasticity and memory formation. We demonstrate that loss of RyR2 in pyramidal neurons of the hippocampus impairs maintenance and activity-evoked structural plasticity of dendritic spines during memory acquisition. Furthermore, post-developmental deletion of RyR2 causes loss of excitatory synapses, dendritic sparsification, overcompensatory excitability, network hyperactivity and disruption of spatially tuned place cells. Altogether, our data underpin RyR2 as a link between spine remodeling, circuitry dysfunction and memory acquisition, which closely resemble pathological mechanisms observed in neurodegenerative disorders.
Collapse
Affiliation(s)
- Fabio Bertan
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Lena Wischhof
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | - Manuel Mittag
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Dennis Dalügge
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Elena Marcello
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Monica Di Luca
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Martin Fuhrmann
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Stefan Remy
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Department of Cellular Neuroscience, Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Daniele Bano
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | |
Collapse
|
40
|
Starikov L, Kottmann AH. Diminished Ventral Oligodendrocyte Precursor Generation Results in the Subsequent Over-production of Dorsal Oligodendrocyte Precursors of Aberrant Morphology and Function. Neuroscience 2020; 450:15-28. [PMID: 32450295 DOI: 10.1016/j.neuroscience.2020.05.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 12/28/2022]
Abstract
Oligodendrocyte precursor cells (OPCs) arise sequentially first from a ventral and then from a dorsal precursor domain at the end of neurogenesis during spinal cord development. Whether the sequential production of OPCs is of physiological significance has not been examined. Here we show that ablating Shh signaling from nascent ventricular zone derivatives and partially from the floor plate results in a severe diminishment of ventral derived OPCs but normal numbers of motor neurons in the postnatal spinal cord. In the absence of ventral vOPCs, dorsal dOPCs populate the entire spinal cord resulting in an increased OPC density in the ventral horns. These OPCs take on an altered morphology, do not participate in the removal of excitatory vGlut1 synapses from injured motor neurons, and exhibit morphological features similar to those found in the vicinity of motor neurons in the SOD1 mouse model of Amyotrophic Lateral Sclerosis (ALS). Our data indicate that vOPCs prevent dOPCs from invading ventral spinal cord laminae and suggest that vOPCs have a unique ability to communicate with injured motor neurons.
Collapse
Affiliation(s)
- Lev Starikov
- City University of New York School of Medicine (CSOM) at City College of New York, Dept. of Molecular, Cellular and Biomedical Sciences, New York City, NY 10031, USA; City University of New York Graduate Center, Molecular, Cellular and Developmental Subprogram, New York City, NY 10016, USA
| | - Andreas H Kottmann
- City University of New York School of Medicine (CSOM) at City College of New York, Dept. of Molecular, Cellular and Biomedical Sciences, New York City, NY 10031, USA; City University of New York Graduate Center, Molecular, Cellular and Developmental Subprogram, New York City, NY 10016, USA.
| |
Collapse
|
41
|
Kermen F, Lal P, Faturos NG, Yaksi E. Interhemispheric connections between olfactory bulbs improve odor detection. PLoS Biol 2020; 18:e3000701. [PMID: 32310946 PMCID: PMC7192517 DOI: 10.1371/journal.pbio.3000701] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 04/30/2020] [Accepted: 04/02/2020] [Indexed: 01/06/2023] Open
Abstract
Interhemispheric connections enable interaction and integration of sensory information in bilaterian nervous systems and are thought to optimize sensory computations. However, the cellular and spatial organization of interhemispheric networks and the computational properties they mediate in vertebrates are still poorly understood. Thus, it remains unclear to what extent the connectivity between left and right brain hemispheres participates in sensory processing. Here, we show that the zebrafish olfactory bulbs (OBs) receive direct interhemispheric projections from their contralateral counterparts in addition to top-down inputs from the contralateral zebrafish homolog of olfactory cortex. The direct interhemispheric projections between the OBs reach peripheral layers of the contralateral OB and retain a precise topographic organization, which directly connects similarly tuned olfactory glomeruli across hemispheres. In contrast, interhemispheric top-down inputs consist of diffuse projections that broadly innervate the inhibitory granule cell layer. Jointly, these interhemispheric connections elicit a balance of topographically organized excitation and nontopographic inhibition on the contralateral OB and modulate odor responses. We show that the interhemispheric connections in the olfactory system enable the modulation of odor response and contribute to a small but significant improvement in the detection of a reproductive pheromone when presented together with complex olfactory cues by potentiating the response of the pheromone selective neurons. Taken together, our data show a previously unknown function for an interhemispheric connection between chemosensory maps of the olfactory system. Interhemispheric connections enable interaction and integration of sensory information in bilaterian nervous systems and are thought to optimize sensory computations. This study shows that interhemispheric olfactory connections in the zebrafish brain improve the detection of a reproductive pheromone within a noisy odor background.
Collapse
Affiliation(s)
- Florence Kermen
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Neuro-Electronics Research Flanders, Leuven, Belgium
- Department of Biology, Faculty of Natural Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- * E-mail: (FK); (EY)
| | - Pradeep Lal
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Nicholas G. Faturos
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Emre Yaksi
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Neuro-Electronics Research Flanders, Leuven, Belgium
- * E-mail: (FK); (EY)
| |
Collapse
|
42
|
Bates AS, Manton JD, Jagannathan SR, Costa M, Schlegel P, Rohlfing T, Jefferis GSXE. The natverse, a versatile toolbox for combining and analysing neuroanatomical data. eLife 2020; 9:e53350. [PMID: 32286229 PMCID: PMC7242028 DOI: 10.7554/elife.53350] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 04/11/2020] [Indexed: 11/18/2022] Open
Abstract
To analyse neuron data at scale, neuroscientists expend substantial effort reading documentation, installing dependencies and moving between analysis and visualisation environments. To facilitate this, we have developed a suite of interoperable open-source R packages called the natverse. The natverse allows users to read local and remote data, perform popular analyses including visualisation and clustering and graph-theoretic analysis of neuronal branching. Unlike most tools, the natverse enables comparison across many neurons of morphology and connectivity after imaging or co-registration within a common template space. The natverse also enables transformations between different template spaces and imaging modalities. We demonstrate tools that integrate the vast majority of Drosophila neuroanatomical light microscopy and electron microscopy connectomic datasets. The natverse is an easy-to-use environment for neuroscientists to solve complex, large-scale analysis challenges as well as an open platform to create new code and packages to share with the community.
Collapse
Affiliation(s)
| | - James D Manton
- Neurobiology Division, MRC Laboratory of Molecular BiologyCambridgeUnited Kingdom
| | - Sridhar R Jagannathan
- Drosophila Connectomics Group, Department of Zoology, University of CambridgeCambridgeUnited Kingdom
| | - Marta Costa
- Neurobiology Division, MRC Laboratory of Molecular BiologyCambridgeUnited Kingdom
- Drosophila Connectomics Group, Department of Zoology, University of CambridgeCambridgeUnited Kingdom
| | - Philipp Schlegel
- Neurobiology Division, MRC Laboratory of Molecular BiologyCambridgeUnited Kingdom
- Drosophila Connectomics Group, Department of Zoology, University of CambridgeCambridgeUnited Kingdom
| | - Torsten Rohlfing
- SRI International, Neuroscience Program, Center for Health SciencesMenlo ParkUnited States
| | - Gregory SXE Jefferis
- Neurobiology Division, MRC Laboratory of Molecular BiologyCambridgeUnited Kingdom
- Drosophila Connectomics Group, Department of Zoology, University of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
43
|
Gradual wiring of olfactory input to amygdala feedback circuits. Sci Rep 2020; 10:5871. [PMID: 32245993 PMCID: PMC7125095 DOI: 10.1038/s41598-020-62457-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 03/11/2020] [Indexed: 11/08/2022] Open
Abstract
The amygdala facilitates odor driven behavioral responses by enhancing the saliency of olfactory signals. Before this processing, olfactory input is refined through the feedback provided by amygdala corticofugal projection (ACPs). Although the saliency of odor signals is subject to developmental changes, the stage at which this cortical feedback first occurs is not known. Using optogenetically-assisted intracellular recordings of the mouse cortical amygdala, we identified changes in the electrophysiological properties of ACPs at different developmental stages. These were consistent with a decrease in neuronal excitability and an increase in the amount of incoming accessory olfactory bulb (AOB) inputs, as confirmed by estimates of release probability, quantal size and contact number at the AOB-to-ACP synapse. Moreover, the proportion of ACPs activated in response to odors was dependent on the stage of development as revealed by c-Fos expression analysis. These results update standard accounts of how the amygdala processes social signals by emphasizing the occurrence of critical periods in the development of its sensory gating functions.
Collapse
|
44
|
Kuhn KD, Edamura K, Bhatia N, Cheng I, Clark SA, Haynes CV, Heffner DL, Kabir F, Velasquez J, Spano AJ, Deppmann CD, Keeler AB. Molecular dissection of TNFR-TNFα bidirectional signaling reveals both cooperative and antagonistic interactions with p75 neurotrophic factor receptor in axon patterning. Mol Cell Neurosci 2020; 103:103467. [PMID: 32004684 PMCID: PMC7682658 DOI: 10.1016/j.mcn.2020.103467] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/18/2019] [Accepted: 01/13/2020] [Indexed: 11/25/2022] Open
Abstract
During neural development, complex organisms rely on progressive and regressive events whereby axons, synapses, and neurons are overproduced followed by selective elimination of a portion of these components. Tumor necrosis factor α (TNFα) together with its cognate receptor (Tumor necrosis factor receptor 1; TNFR1) have been shown to play both regressive (i.e. forward signaling from the receptor) and progressive (i.e. reverse signaling from the ligand) roles in sympathetic neuron development. In contrast, a paralog of TNFR1, p75 neurotrophic factor receptor (p75NTR) promotes mainly regressive developmental events in sympathetic neurons. Here we examine the interplay between these paralogous receptors in the regulation of axon branch elimination and arborization. We confirm previous reports that these TNFR1 family members are individually capable of promoting ligand-dependent suppression of axon growth and branching. Remarkably, p75NTR and TNFR1 physically interact and p75NTR requires TNFR1 for ligand-dependent axon suppression of axon branching but not vice versa. We also find that p75NTR forward signaling and TNFα reverse signaling are functionally antagonistic. Finally, we find that TNFα reverse signaling is necessary for nerve growth factor (NGF) dependent axon growth. Taken together these findings demonstrate several levels of synergistic and antagonistic interactions using very few signaling pathways and that the balance of these synergizing and opposing signals act to ensure proper axon growth and patterning.
Collapse
Affiliation(s)
- K D Kuhn
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - K Edamura
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - N Bhatia
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - I Cheng
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22903, USA
| | - S A Clark
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22903, USA
| | - C V Haynes
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - D L Heffner
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - F Kabir
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - J Velasquez
- Blue Ridge Virtual Governor's School, Palmyra, VA 22963, USA
| | - A J Spano
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - C D Deppmann
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22903, USA; Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22903, USA; Department of Cell Biology, University of Virginia, Charlottesville, VA 22903, USA.
| | - A B Keeler
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA.
| |
Collapse
|
45
|
Raghuram V, Werginz P, Fried SI. Scaling of the AIS and Somatodendritic Compartments in α S RGCs. Front Cell Neurosci 2019; 13:436. [PMID: 31611777 PMCID: PMC6777007 DOI: 10.3389/fncel.2019.00436] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 09/13/2019] [Indexed: 11/21/2022] Open
Abstract
The anatomical properties of the axon initial segment (AIS) are tailored in certain types of CNS neurons to help optimize different aspects of neuronal function. Here, we questioned whether the AISs of retinal ganglion cells (RGC) were similarly customized, and if so, whether they supported specific RGC functions. To explore this, we measured the AIS properties in alpha sustained RGCs (α S RGCs) of mouse; α S RGCs sizes vary systematically along the nasal temporal axis of the retina, making these cells an attractive population with which to study potential correlations between AIS properties and cell size. Measurements of AIS length as well as distance from the soma revealed that both were scaled to cell size, i.e., cells with large dendritic fields had long AISs that were relatively far from the soma. Within the AIS, the percentage of Na v 1.6 voltage-gated sodium channels remained highly consistent, regardless of cell size or other AIS properties. Although ON RGCs were slightly larger than OFF cells at any given location of the retina, the level of scaling and relative distribution of voltage-gated sodium channels were highly similar. Computational modeling revealed that AIS scaling influenced spiking thresholds, spike rate as well as the kinetics of individual action potentials, Interestingly, the effect of individual features of the AIS varied for different neuronal functions, e.g., AIS length had a larger effect on the efficacy by which the AIS initiated spike triggered the somatic spike than it did on repetitive spiking. The polarity of the effect varied for different properties, i.e., increases to soma size increased spike threshold while increases to AIS length decreased threshold. Thus, variations in the relative level of scaling for individual components could fine tune threshold or other neuronal functions. Light responses were highly consistent across the full range of cell sizes suggesting that scaling may post-synaptically shape response stability, e.g., in addition to several well-known pre-synaptic contributors.
Collapse
Affiliation(s)
- Vineeth Raghuram
- Rehabilitation Research & Development Service, Boston VA Healthcare System, Boston, MA, United States
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Paul Werginz
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Institute for Analysis and Scientific Computing, Vienna University of Technology, Vienna, Austria
| | - Shelley I. Fried
- Rehabilitation Research & Development Service, Boston VA Healthcare System, Boston, MA, United States
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
46
|
Wang CF, Zhao CC, Liu WL, Huang XJ, Deng YF, Jiang JY, Li WP. Depletion of Microglia Attenuates Dendritic Spine Loss and Neuronal Apoptosis in the Acute Stage of Moderate Traumatic Brain Injury in Mice. J Neurotrauma 2019; 37:43-54. [PMID: 31397209 DOI: 10.1089/neu.2019.6460] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Microglia are the primary immune cells in the central nervous system and undergo significant morphological and transcriptional changes after traumatic brain injury (TBI). However, their exact contribution to the pathogenesis of TBI is still debated and remains to be elucidated. In the present study, thy-1 GFP mice received a colony-stimulating factor 1 receptor inhibitor (PLX3397) for 21 consecutive days, then were subjected to moderate fluid percussion injury (FPI). Brain samples were collected at 1 day and 3 days after FPI for flow cytometry analysis, immunofluorescence, dendrite spine quantification, terminal deoxynucleotidyl transferase dUTP nick end labeling assay, and Western blot. We found that PLX3397 treatment significantly attenuated the percentages of resident microglia and infiltrated immune cells. Depletion of microglia promoted neurite outgrowth, preserved dendritic spines and reduced total brain cell and neuronal apoptosis after FPI, which was accompanied by decreased the protein levels of endoplasmic reticulum stress marker proteins, C/EBP-homologous protein and inositol-requiring kinase 1α. Taken together, these findings suggest that microglial depletion may exert beneficial effects in the acute stage of FPI.
Collapse
Affiliation(s)
- Chuan-Fang Wang
- Brain Center, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Second People's Hospital, Shenzhen, Guangdong, China
| | - Cheng-Cheng Zhao
- Department of Neurosurgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wen-Lan Liu
- Brain Center, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Second People's Hospital, Shenzhen, Guangdong, China
| | - Xian-Jian Huang
- Brain Center, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Second People's Hospital, Shenzhen, Guangdong, China
| | - Yue-Fei Deng
- Department of Neurosurgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ji-Yao Jiang
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei-Ping Li
- Brain Center, Shenzhen Key Laboratory of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Second People's Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
47
|
Everaerts S, McDonough JE, Verleden SE, Josipovic I, Boone M, Dubbeldam A, Mathyssen C, Serré J, Dupont LJ, Gayan-Ramirez G, Verschakelen J, Hogg JC, Verleden GM, Vanaudenaerde BM, Janssens W. Airway morphometry in COPD with bronchiectasis: a view on all airway generations. Eur Respir J 2019; 54:13993003.02166-2018. [DOI: 10.1183/13993003.02166-2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 08/03/2019] [Indexed: 11/05/2022]
Abstract
The pathophysiological processes underlying bronchiectasis in chronic obstructive pulmonary disease (COPD) are not understood. In COPD, both small and large airways are progressively lost. It is currently not known to what extent the different airway generations of patients with COPD and bronchiectasis are involved.COPD explant lungs with bronchiectasis were compared to COPD explant lungs without bronchiectasis and unused donor lungs as controls. In order to investigate all airway generations, a multimodal imaging approach using different resolutions was conducted. Per group, five lungs were frozen (n=15) and underwent computed tomography (CT) imaging for large airway evaluation, with four tissue cores per lung imaged for measurements of the terminal bronchioles. Two additional lungs per group (n=6) were air-dried for lobar microCT images that allow airway segmentation and three-dimensional quantification of the complete airway tree.COPD lungs with bronchiectasis had significantly more airways compared to COPD lungs without bronchiectasis (p<0.001), with large airway numbers similar to control lungs. This difference was present in both upper and lower lobes. Lack of tapering was present (p=0.010) and larger diameters were demonstrated in lower lobes with bronchiectasis (p=0.010). MicroCT analysis of tissue cores showed similar reductions of tissue percentage, surface density and number of terminal bronchioles in both COPD groups compared to control lungs.Although terminal bronchioles were equally reduced in COPD lungs with and without bronchiectasis, significantly more large and small airways were found in COPD lungs with bronchiectasis.
Collapse
|
48
|
FMST: an Automatic Neuron Tracing Method Based on Fast Marching and Minimum Spanning Tree. Neuroinformatics 2019; 17:185-196. [PMID: 30039210 DOI: 10.1007/s12021-018-9392-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Neuron reconstruction is an important technique in computational neuroscience. Although there are many reconstruction algorithms, few can generate robust results. In this paper, we propose a reconstruction algorithm called fast marching spanning tree (FMST). FMST is based on a minimum spanning tree method (MST) and improve its performance in two aspects: faster implementation and no loss of small branches. The contributions of the proposed method are as follows. Firstly, the Euclidean distance weight of edges in MST is improved to be a more reasonable value, which is related to the probability of the existence of an edge. Secondly, a strategy of pruning nodes is presented, which is based on the radius of a node's inscribed ball. Thirdly, separate branches of broken neuron reconstructions can be merged into a single tree. FMST and many other state of the art reconstruction methods were implemented on two datasets: 120 Drosophila neurons and 163 neurons with gold standard reconstructions. Qualitative and quantitative analysis on experimental results demonstrates that the performance of FMST is good compared with many existing methods. Especially, on the 91 fruitfly neurons with gold standard and evaluated by five metrics, FMST is one of two methods with best performance among all 27 state of the art reconstruction methods. FMST is a good and practicable neuron reconstruction algorithm, and can be implemented in Vaa3D platform as a neuron tracing plugin.
Collapse
|
49
|
Liu M, Chen W, Wang C, Peng H. A Multiscale Ray-Shooting Model for Termination Detection of Tree-Like Structures in Biomedical Images. IEEE TRANSACTIONS ON MEDICAL IMAGING 2019; 38:1923-1934. [PMID: 30668496 DOI: 10.1109/tmi.2019.2893117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Digital reconstruction (tracing) of tree-like structures, such as neurons, retinal blood vessels, and bronchi, from volumetric images and 2D images is very important to biomedical research. Many existing reconstruction algorithms rely on a set of good seed points. The 2D or 3D terminations are good candidates for such seed points. In this paper, we propose an automatic method to detect terminations for tree-like structures based on a multiscale ray-shooting model and a termination visual prior. The multiscale ray-shooting model detects 2D terminations by extracting and analyzing the multiscale intensity distribution features around a termination candidate. The range of scale is adaptively determined according to the local neurite diameter estimated by the Rayburst sampling algorithm in combination with the gray-weighted distance transform. The termination visual prior is based on a key observation-when observing a 3D termination from three orthogonal directions without occlusion, we can recognize it in at least two views. Using this prior with the multiscale ray-shooting model, we can detect 3D terminations with high accuracies. Experiments on 3D neuron image stacks, 2D neuron images, 3D bronchus image stacks, and 2D retinal blood vessel images exhibit average precision and recall rates of 87.50% and 90.54%. The experimental results confirm that the proposed method outperforms other the state-of-the-art termination detection methods.
Collapse
|
50
|
Zhu Y, Liu B, Zheng X, Wu J, Chen S, Chen Z, Chen T, Huang Z, Lei W. Partial decortication ameliorates dopamine depletion‑induced striatal neuron lesions in rats. Int J Mol Med 2019; 44:1414-1424. [PMID: 31364729 PMCID: PMC6713435 DOI: 10.3892/ijmm.2019.4288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 06/27/2019] [Indexed: 12/25/2022] Open
Abstract
The balance between glutamate (cortex and thalamus) and dopamine (substantia nigra) inputs on striatal neurons is of vital importance. Dopamine deficiency, which breaks this balance and leads to the domination of cortical glutamatergic inputs, plays an important role in Parkinson's disease (PD). However, the exact impact on striatal neurons has not been fully clarified. Thus, the present study aimed to characterize the influence of corticostriatal glutamatergic inputs on striatal neurons after decortication due to dopamine depletion in rats. 6-Hydroxydopamine was injected into the right medial forebrain bundle to induce dopamine depletion, and/or ibotenic acid into the primary motor cortex to induce decortication. Subsequently, the grip strength test and Morris water maze task indicated that decortication significantly shortened the hang time and the latency that had been increased in the rats subjected to dopamine depletion. Golgi staining and electron microscopy analysis showed that the total dendritic length and dendritic spine density of the striatal neurons were decreased in the dopamine-depleted rats, whereas decortication alleviated this damage. Immunohistochemistry analysis demonstrated that decortication decreased the number of caspase-3-positive neurons in the dopamine-depleted rats. Moreover, reverse transcription-quantitative PCR and western blot analyses showed that decortication offset the upregulation of caspase-3 at both the protein and mRNA levels in the dopamine-depleted rats. In conclusion, the present study demonstrated that a relative excess of cortical glutamate inputs had a substantial impact on the pathological processes of striatal neuron lesions in PD.
Collapse
Affiliation(s)
- Yaofeng Zhu
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Bingbing Liu
- Department of Anesthesiology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, P.R. China
| | - Xuefeng Zheng
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Jiajia Wu
- Periodical Center, The Third Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Si Chen
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong 510006, P.R. China
| | - Zhi Chen
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Tao Chen
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Ziyun Huang
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Wanlong Lei
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|