1
|
Martinez-Lozada Z, Guillem AM, Song I, Gonzalez MV, Takano H, Parikh E, Rothstein JD, Putt ME, Robinson MB. Identification of a Subpopulation of Astrocyte Progenitor Cells in the Neonatal Subventricular Zone: Evidence that Migration is Regulated by Glutamate Signaling. Neurochem Res 2025; 50:77. [PMID: 39789409 PMCID: PMC11717811 DOI: 10.1007/s11064-024-04326-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025]
Abstract
In mice engineered to express enhanced green fluorescent protein (eGFP) under the control of the entire glutamate transporter 1 (GLT1) gene, eGFP is found in all 'adult' cortical astrocytes. However, when 8.3 kilobases of the human GLT1/EAAT2 promoter is used to control expression of tdTomato (tdT), tdT is only found in a subpopulation of these eGFP-expressing astrocytes. The eGFP mice have been used to define mechanisms of transcriptional regulation using astrocytes cultured from cortex of 1-3 day old mice. Using the same cultures, we were never able to induce tdT+ expression. We hypothesized that these cells might not have migrated into the cortex by this age. In this study, we characterized the ontogeny of tdT+ cells, performed single-cell RNA sequencing (scRNA-seq), and tracked their migration in organotypic slice cultures. At postnatal day (PND) 1, tdT+ cells were observed in the subventricular zone and striatum but not in the cortex, and they did not express eGFP. At PND7, tdT+ cells begin to appear in the cortex with their numbers increasing with age. At PND1, scRNA-seq demonstrates that the tdT+ cells are molecularly heterogeneous, with a subpopulation expressing astrocytic markers, subsequently validated with immunofluorescence. In organotypic slices, tdT+ cells migrate into the cortex, and after 7 days they express GLT1, NF1A, and eGFP. An ionotropic glutamate receptor (iGluR) antagonist reduced by 50% the distance tdT+ cells migrate from the subventricular zone into the cortex. The pan-glutamate transport inhibitor, TFB-TBOA, increased, by sixfold, the number of tdT+ cells in the cortex. In conclusion, although tdT is expressed by non-glial cells at PND1, it is also expressed by glial progenitors that migrate into the cortex postnatally. Using this fluorescent labeling, we provide novel evidence that glutamate signaling contributes to the control of glial precursor migration.
Collapse
Affiliation(s)
- Zila Martinez-Lozada
- Departments of Pediatrics and Systems Pharmacology & Translational Therapeutics, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, 19104-4318, USA.
- Department of Neuroscience, College of Psychology and Neuroscience, Nova Southeastern University, Fort Lauderdale, FL, 33328, USA.
| | - Alain M Guillem
- Departments of Pediatrics and Systems Pharmacology & Translational Therapeutics, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, 19104-4318, USA
| | - Isabella Song
- Departments of Pediatrics and Systems Pharmacology & Translational Therapeutics, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, 19104-4318, USA
| | - Michael V Gonzalez
- Center for Cytokine Storm Treatment & Laboratory, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hajime Takano
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Esha Parikh
- Departments of Pediatrics and Systems Pharmacology & Translational Therapeutics, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, 19104-4318, USA
| | - Jeffrey D Rothstein
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Mary E Putt
- Department of Biostatistics, Epidemiology & Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Michael B Robinson
- Departments of Pediatrics and Systems Pharmacology & Translational Therapeutics, The Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, 19104-4318, USA.
| |
Collapse
|
2
|
Lerma-Martin C, Badia-I-Mompel P, Ramirez Flores RO, Sekol P, Schäfer PSL, Riedl CJ, Hofmann A, Thäwel T, Wünnemann F, Ibarra-Arellano MA, Trobisch T, Eisele P, Schapiro D, Haeussler M, Hametner S, Saez-Rodriguez J, Schirmer L. Cell type mapping reveals tissue niches and interactions in subcortical multiple sclerosis lesions. Nat Neurosci 2024; 27:2354-2365. [PMID: 39501036 PMCID: PMC11614744 DOI: 10.1038/s41593-024-01796-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: 10/23/2022] [Accepted: 09/30/2024] [Indexed: 11/08/2024]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system. Inflammation is gradually compartmentalized and restricted to specific tissue niches such as the lesion rim. However, the precise cell type composition of such niches, their interactions and changes between chronic active and inactive stages are incompletely understood. We used single-nucleus and spatial transcriptomics from subcortical MS and corresponding control tissues to map cell types and associated pathways to lesion and nonlesion areas. We identified niches such as perivascular spaces, the inflamed lesion rim or the lesion core that are associated with the glial scar and a cilia-forming astrocyte subtype. Focusing on the inflamed rim of chronic active lesions, we uncovered cell-cell communication events between myeloid, endothelial and glial cell types. Our results provide insight into the cellular composition, multicellular programs and intercellular communication in tissue niches along the conversion from a homeostatic to a dysfunctional state underlying lesion progression in MS.
Collapse
Affiliation(s)
- Celia Lerma-Martin
- Department of Neurology, Division of Neuroimmunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Pau Badia-I-Mompel
- Institute for Computational Biomedicine, Faculty of Medicine, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
- GSK, Cellzome, Heidelberg, Germany
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, UK
| | - Ricardo O Ramirez Flores
- Institute for Computational Biomedicine, Faculty of Medicine, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, UK
| | - Patricia Sekol
- Department of Neurology, Division of Neuroimmunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Philipp S L Schäfer
- Institute for Computational Biomedicine, Faculty of Medicine, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, UK
| | - Christian J Riedl
- Department of Neurology, Division of Neuropathology and Neurochemistry, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Annika Hofmann
- Department of Neurology, Division of Neuroimmunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Thomas Thäwel
- Department of Neurology, Division of Neuroimmunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Florian Wünnemann
- Institute for Computational Biomedicine, Faculty of Medicine, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
| | - Miguel A Ibarra-Arellano
- Institute for Computational Biomedicine, Faculty of Medicine, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
| | - Tim Trobisch
- Department of Neurology, Division of Neuroimmunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Philipp Eisele
- Department of Neurology, Division of Neuroimmunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Mannheim Center for Translational Neuroscience, Medical Faculty, Mannheim Heidelberg University, Mannheim, Germany
| | - Denis Schapiro
- Institute for Computational Biomedicine, Faculty of Medicine, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
- Institute of Pathology, Faculty of Medicine, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany
- Translational Spatial Profiling Center (TSPC), Heidelberg, Germany
| | | | - Simon Hametner
- Department of Neurology, Division of Neuropathology and Neurochemistry, Medical University of Vienna, Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Julio Saez-Rodriguez
- Institute for Computational Biomedicine, Faculty of Medicine, Heidelberg University and Heidelberg University Hospital, Heidelberg, Germany.
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, UK.
| | - Lucas Schirmer
- Department of Neurology, Division of Neuroimmunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- Mannheim Center for Translational Neuroscience, Medical Faculty, Mannheim Heidelberg University, Mannheim, Germany.
- Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
3
|
Nakayama-Kitamura K, Shigemoto-Mogami Y, Piantino M, Naka Y, Yamada A, Kitano S, Furihata T, Matsusaki M, Sato K. Collagen I Microfiber Promotes Brain Capillary Network Formation in Three-Dimensional Blood-Brain Barrier Microphysiological Systems. Biomedicines 2024; 12:2500. [PMID: 39595066 PMCID: PMC11591679 DOI: 10.3390/biomedicines12112500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND The blood-brain barrier (BBB) strictly regulates the penetration of substances into the brain, which, although important for maintaining brain homeostasis, may delay drug development because of the difficulties in predicting pharmacokinetics/pharmacodynamics (PKPD), toxicokinetics/toxicodynamics (TKTD), toxicity, safety, and efficacy in the central nervous system (CNS). Moreover, BBB functional proteins show species differences; therefore, humanized in vitro BBB models are urgently needed to improve the predictability of preclinical studies. Recently, international trends in the 3Rs in animal experiments and the approval of the FDA Modernization Act 2.0 have accelerated the application of microphysiological systems (MPSs) in preclinical studies, and in vitro BBB models have become synonymous with BBB-MPSs. Recently, we developed an industrialized humanized BBB-MPS, BBB-NET. In our previous report, we reproduced transferrin receptor (TfR)-mediated transcytosis with high efficiency and robustness, using hydrogels including fibrin and collagen I microfibers (CMFs). METHODS We investigated how adding CMFs to the fibrin gel benefits BBB-NETs. RESULTS We showed that CMFs accelerate capillary network formation and maturation by promoting astrocyte (AC) survival, and clarified that integrin β1 is involved in the mechanism of CMFs. CONCLUSIONS Our data suggest that the quality control (QC) of CMFs is important for ensuring the stable production of BBB-NETs.
Collapse
Affiliation(s)
- Kimiko Nakayama-Kitamura
- Laboratory of Neuropharmacology, Division of Pharmacology, National Institute of Health Sciences, 3-25-26, Tonomachi, Kawasaki-ku, Kawasaki City 210-9501, Kanagawa, Japan; (K.N.-K.); (Y.S.-M.)
| | - Yukari Shigemoto-Mogami
- Laboratory of Neuropharmacology, Division of Pharmacology, National Institute of Health Sciences, 3-25-26, Tonomachi, Kawasaki-ku, Kawasaki City 210-9501, Kanagawa, Japan; (K.N.-K.); (Y.S.-M.)
| | - Marie Piantino
- Joint Research Laboratory for Social Implementation of Cultured Meat, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan; (M.P.); (M.M.)
| | - Yasuhiro Naka
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan;
| | - Asuka Yamada
- TOPPAN Holdings Inc., TOPPAN Technical Research Institute, 4-2 Takanodaiminami, Sugitomachi, Saitama 345-8508, Saitama, Japan; (A.Y.); (S.K.)
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Shiro Kitano
- TOPPAN Holdings Inc., TOPPAN Technical Research Institute, 4-2 Takanodaiminami, Sugitomachi, Saitama 345-8508, Saitama, Japan; (A.Y.); (S.K.)
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Tomomi Furihata
- Laboratory of Advanced Drug Developmen Sciences, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji 192-0392, Tokyo, Japan;
| | - Michiya Matsusaki
- Joint Research Laboratory for Social Implementation of Cultured Meat, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan; (M.P.); (M.M.)
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan;
| | - Kaoru Sato
- Laboratory of Neuropharmacology, Division of Pharmacology, National Institute of Health Sciences, 3-25-26, Tonomachi, Kawasaki-ku, Kawasaki City 210-9501, Kanagawa, Japan; (K.N.-K.); (Y.S.-M.)
| |
Collapse
|
4
|
Schubert MC, Soyka SJ, Tamimi A, Maus E, Schroers J, Wißmann N, Reyhan E, Tetzlaff SK, Yang Y, Denninger R, Peretzke R, Beretta C, Drumm M, Heuer A, Buchert V, Steffens A, Walshon J, McCortney K, Heiland S, Bendszus M, Neher P, Golebiewska A, Wick W, Winkler F, Breckwoldt MO, Kreshuk A, Kuner T, Horbinski C, Kurz FT, Prevedel R, Venkataramani V. Deep intravital brain tumor imaging enabled by tailored three-photon microscopy and analysis. Nat Commun 2024; 15:7383. [PMID: 39256378 PMCID: PMC11387418 DOI: 10.1038/s41467-024-51432-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 08/07/2024] [Indexed: 09/12/2024] Open
Abstract
Intravital 2P-microscopy enables the longitudinal study of brain tumor biology in superficial mouse cortex layers. Intravital microscopy of the white matter, an important route of glioblastoma invasion and recurrence, has not been feasible, due to low signal-to-noise ratios and insufficient spatiotemporal resolution. Here, we present an intravital microscopy and artificial intelligence-based analysis workflow (Deep3P) that enables longitudinal deep imaging of glioblastoma up to a depth of 1.2 mm. We find that perivascular invasion is the preferred invasion route into the corpus callosum and uncover two vascular mechanisms of glioblastoma migration in the white matter. Furthermore, we observe morphological changes after white matter infiltration, a potential basis of an imaging biomarker during early glioblastoma colonization. Taken together, Deep3P allows for a non-invasive intravital investigation of brain tumor biology and its tumor microenvironment at subcortical depths explored, opening up opportunities for studying the neuroscience of brain tumors and other model systems.
Collapse
Affiliation(s)
- Marc Cicero Schubert
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Stella Judith Soyka
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Amr Tamimi
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Emanuel Maus
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Julian Schroers
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
- German Cancer Research Center (DKFZ), Division of Radiology, Heidelberg, Germany
| | - Niklas Wißmann
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Ekin Reyhan
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Svenja Kristin Tetzlaff
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Yvonne Yang
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Robert Denninger
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Robin Peretzke
- Division of Medical Image Computing (MIC), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Carlo Beretta
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Michael Drumm
- Department of Neurological Surgery, Northwestern University, Chicago, IL, USA
| | - Alina Heuer
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Verena Buchert
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Alicia Steffens
- Department of Neurological Surgery, Northwestern University, Chicago, IL, USA
| | - Jordain Walshon
- Department of Neurological Surgery, Northwestern University, Chicago, IL, USA
| | - Kathleen McCortney
- Department of Neurological Surgery, Northwestern University, Chicago, IL, USA
| | - Sabine Heiland
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Martin Bendszus
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Peter Neher
- Division of Medical Image Computing (MIC), German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Anna Golebiewska
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health, 1526, Luxembourg, Luxembourg
| | - Wolfgang Wick
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frank Winkler
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael O Breckwoldt
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Anna Kreshuk
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Thomas Kuner
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Craig Horbinski
- Department of Neurological Surgery, Northwestern University, Chicago, IL, USA
- Department of Pathology, Northwestern University, Chicago, IL, USA
| | - Felix Tobias Kurz
- German Cancer Research Center (DKFZ), Division of Radiology, Heidelberg, Germany
- Department of Neuroradiology, University Hospital Heidelberg, Heidelberg, Germany
- Division of Neuroradiology, Geneva University Hospitals, Geneva, Switzerland
| | - Robert Prevedel
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.
- Developmental Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
- Epigenetics and Neurobiology Unit, European Molecular Biology Laboratory, Rome, Italy.
- Molecular Medicine Partnership Unit (MMPU), European Molecular Biology Laboratory, Heidelberg, Germany.
- Interdisciplinary Center of Neurosciences, Heidelberg University, Heidelberg, Germany.
| | - Varun Venkataramani
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany.
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
5
|
Cui Y, Rolova T, Fagerholm SC. The role of integrins in brain health and neurodegenerative diseases. Eur J Cell Biol 2024; 103:151441. [PMID: 39002282 DOI: 10.1016/j.ejcb.2024.151441] [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: 04/29/2024] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 07/15/2024] Open
Abstract
Integrins are heterodimeric membrane proteins expressed on the surface of most cells. They mediate adhesion and signaling processes relevant for a wealth of physiological processes, including nervous system development and function. Interestingly, integrins are also recognized therapeutic targets for inflammatory diseases, such as multiple sclerosis. Here, we discuss the role of integrins in brain development and function, as well as in neurodegenerative diseases affecting the brain (Alzheimer's disease, multiple sclerosis, stroke). Furthermore, we discuss therapeutic targeting of these adhesion receptors in inflammatory diseases of the brain.
Collapse
Affiliation(s)
- Yunhao Cui
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki 00790, Finland
| | - Taisia Rolova
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki 00290, Finland
| | - Susanna C Fagerholm
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki 00790, Finland.
| |
Collapse
|
6
|
Lozano Casasbuenas D, Kortebi I, Gora C, Scott EY, Gomes C, Oliveira MS, Sharma T, Daniele E, Olfat A, Gibbs R, Yuzwa SA, Gilbert EA, Küry P, Wheeler AR, Lévesque M, Faiz M. The laminar position, morphology, and gene expression profiles of cortical astrocytes are influenced by time of birth from ventricular/subventricular progenitors. Glia 2024; 72:1693-1706. [PMID: 38852127 DOI: 10.1002/glia.24578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 05/06/2024] [Accepted: 05/27/2024] [Indexed: 06/10/2024]
Abstract
Astrocytes that reside in superficial (SL) and deep cortical layers have distinct molecular profiles and morphologies, which may underlie specific functions. Here, we demonstrate that the production of SL and deep layer (DL) astrocyte populations from neural progenitor cells in the mouse is temporally regulated. Lineage tracking following in utero and postnatal electroporation with PiggyBac (PB) EGFP and birth dating with EdU and FlashTag, showed that apical progenitors produce astrocytes during late embryogenesis (E16.5) that are biased to the SL, while postnatally labeled (P0) astrocytes are biased to the DL. In contrast, astrocytes born during the predominantly neurogenic window (E14.5) showed a random distribution in the SL and DL. Of interest, E13.5 astrocytes birth dated at E13.5 with EdU showed a lower layer bias, while FT labeling of apical progenitors showed no bias. Finally, examination of the morphologies of "biased" E16.5- and P0-labeled astrocytes demonstrated that E16.5-labeled astrocytes exhibit different morphologies in different layers, while P0-labeled astrocytes do not. Differences based on time of birth are also observed in the molecular profiles of E16.5 versus P0-labeled astrocytes. Altogether, these results suggest that the morphological, molecular, and positional diversity of cortical astrocytes is related to their time of birth from ventricular/subventricular zone progenitors.
Collapse
Affiliation(s)
- Daniela Lozano Casasbuenas
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Ines Kortebi
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Charles Gora
- Department of Psychiatry and Neurosciences, Université Laval, Québec, Canada; CERVO Brain Research Center, Québec, Canada
| | - Erica Y Scott
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Celeste Gomes
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Markley Silva Oliveira
- Neuroregeneration Laboratory, Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Tanvi Sharma
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Emerson Daniele
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Arman Olfat
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Rachel Gibbs
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Scott A Yuzwa
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Emily A Gilbert
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Patrick Küry
- Neuroregeneration Laboratory, Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
- Department of Neurology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland
| | - Aaron R Wheeler
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Martin Lévesque
- Department of Psychiatry and Neurosciences, Université Laval, Québec, Canada; CERVO Brain Research Center, Québec, Canada
| | - Maryam Faiz
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Herwerth M, Wyss MT, Schmid NB, Condrau J, Ravotto L, Mateos Melero JM, Kaech A, Bredell G, Thomas C, Stadelmann C, Misgeld T, Bennett JL, Saab AS, Jessberger S, Weber B. Astrocytes adopt a progenitor-like migratory strategy for regeneration in adult brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.18.594292. [PMID: 38798654 PMCID: PMC11118580 DOI: 10.1101/2024.05.18.594292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Mature astrocytes become activated upon non-specific tissue damage and contribute to glial scar formation. Proliferation and migration of adult reactive astrocytes after injury is considered very limited. However, the regenerative behavior of individual astrocytes following selective astroglial loss, as seen in astrocytopathies, such as neuromyelitis optica spectrum disorder, remains unexplored. Here, we performed longitudinal in vivo imaging of cortical astrocytes after focal astrocyte ablation in mice. We discovered that perilesional astrocytes develop a remarkable plasticity for efficient lesion repopulation. A subset of mature astrocytes transforms into reactive progenitor-like (REPL) astrocytes that not only undergo multiple asymmetric divisions but also remain in a multinucleated interstage. This regenerative response facilitates efficient migration of newly formed daughter cell nuclei towards unoccupied astrocyte territories. Our findings define the cellular principles of astrocyte plasticity upon focal lesion, unravelling the REPL phenotype as a fundamental regenerative strategy of mature astrocytes to restore astrocytic networks in the adult mammalian brain. Promoting this regenerative phenotype bears therapeutic potential for neurological conditions involving glial dysfunction.
Collapse
|
8
|
Miyajima M, Tabata H, Nakajima K. Migratory mode transition of astrocyte progenitors in the cerebral cortex: an intrinsic or extrinsic cell process? Neural Regen Res 2024; 19:471-472. [PMID: 37721258 PMCID: PMC10581590 DOI: 10.4103/1673-5374.380886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/23/2023] [Accepted: 06/09/2023] [Indexed: 09/19/2023] Open
Affiliation(s)
- Michio Miyajima
- Department of Anatomy, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Hidenori Tabata
- Department of Anatomy, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai, Aichi, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
9
|
Shvedov NR, Analoui S, Dafalias T, Bedell BL, Gardner TJ, Scott BB. In vivo imaging in transgenic songbirds reveals superdiffusive neuron migration in the adult brain. Cell Rep 2024; 43:113759. [PMID: 38345898 DOI: 10.1016/j.celrep.2024.113759] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 12/01/2023] [Accepted: 01/23/2024] [Indexed: 03/02/2024] Open
Abstract
Neuron migration is a key phase of neurogenesis, critical for the assembly and function of neuronal circuits. In songbirds, this process continues throughout life, but how these newborn neurons disperse through the adult brain is unclear. We address this question using in vivo two-photon imaging in transgenic zebra finches that express GFP in young neurons and other cell types. In juvenile and adult birds, migratory cells are present at a high density, travel in all directions, and make frequent course changes. Notably, these dynamic migration patterns are well fit by a superdiffusive model. Simulations reveal that these superdiffusive dynamics are sufficient to disperse new neurons throughout the song nucleus HVC. These results suggest that superdiffusive migration may underlie the formation and maintenance of nuclear brain structures in the postnatal brain and indicate that transgenic songbirds are a useful resource for future studies into the mechanisms of adult neurogenesis.
Collapse
Affiliation(s)
- Naomi R Shvedov
- Graduate Program for Neuroscience, Boston University, Boston, MA 02215, USA
| | - Sina Analoui
- Department of Psychological and Brain Sciences, Boston University, Boston, MA 02215, USA
| | - Theresia Dafalias
- Graduate Program for Neuroscience, Boston University, Boston, MA 02215, USA
| | - Brooke L Bedell
- Department of Psychological and Brain Sciences, Boston University, Boston, MA 02215, USA
| | - Timothy J Gardner
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR 97403, USA
| | - Benjamin B Scott
- Department of Psychological and Brain Sciences, Boston University, Boston, MA 02215, USA; Neurophotonics Center, Photonics Center, and Center for Systems Neuroscience, Boston University, Boston, MA 02215, USA.
| |
Collapse
|
10
|
Shin M, Kosodo Y. Brain Sample Preparation After Performing in Utero Electroporation to Proliferating and Differentiated Cells in the Developing Mouse Neocortex. Methods Mol Biol 2024; 2794:187-200. [PMID: 38630230 DOI: 10.1007/978-1-0716-3810-1_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
In utero electroporation (IUE) enables labeling and manipulating specific types of cells by introducing DNA plasmids with desired promoters. After the surgery, mouse brains are fixed at any stage and analyzed after staining using specific antibodies. Here, we describe the flow of the IUE experiment from the preparation to microscopic observations.
Collapse
Affiliation(s)
- Minkyung Shin
- Korea Brain Research Institute (KBRI), Daegu, Republic of Korea
| | - Yoichi Kosodo
- Korea Brain Research Institute (KBRI), Daegu, Republic of Korea.
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea.
| |
Collapse
|
11
|
Nishikawa M, Nagata KI, Tabata H. Live Imaging of Migrating Neurons and Glial Progenitors Visualized by in Utero Electroporation. Methods Mol Biol 2024; 2794:201-209. [PMID: 38630231 DOI: 10.1007/978-1-0716-3810-1_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
During cortical development, both neurons and glial cells are generated in the germinal zone near the lateral ventricle, migrate in the correct direction, and settle in their appropriate locations. This developmental process can be clearly visualized by introducing fluorescent protein-expression vectors via in utero electroporation. In this chapter, we describe labeling methods for migrating neurons and glial progenitors, as well as methods for slice culture, and time-lapse imaging.
Collapse
Affiliation(s)
- Masashi Nishikawa
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai, Aichi, Japan
| | - Koh-Ichi Nagata
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai, Aichi, Japan
- Department of Neurochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hidenori Tabata
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, Kasugai, Aichi, Japan.
| |
Collapse
|
12
|
Yang C, Shitamukai A, Yang S, Kawaguchi A. Advanced Techniques Using In Vivo Electroporation to Study the Molecular Mechanisms of Cerebral Development Disorders. Int J Mol Sci 2023; 24:14128. [PMID: 37762431 PMCID: PMC10531473 DOI: 10.3390/ijms241814128] [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: 08/10/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
The mammalian cerebral cortex undergoes a strictly regulated developmental process. Detailed in situ visualizations, imaging of these dynamic processes, and in vivo functional gene studies significantly enhance our understanding of brain development and related disorders. This review introduces basic techniques and recent advancements in in vivo electroporation for investigating the molecular mechanisms underlying cerebral diseases. In utero electroporation (IUE) is extensively used to visualize and modify these processes, including the forced expression of pathological mutants in human diseases; thus, this method can be used to establish animal disease models. The advent of advanced techniques, such as genome editing, including de novo knockout, knock-in, epigenetic editing, and spatiotemporal gene regulation, has further expanded our list of investigative tools. These tools include the iON expression switch for the precise control of timing and copy numbers of exogenous genes and TEMPO for investigating the temporal effects of genes. We also introduce the iGONAD method, an improved genome editing via oviductal nucleic acid delivery approach, as a novel genome-editing technique that has accelerated brain development exploration. These advanced in vivo electroporation methods are expected to provide valuable insights into pathological conditions associated with human brain disorders.
Collapse
Affiliation(s)
- Chen Yang
- Human Anatomy and Histology and Embryology, School of Basic Medicine, Harbin Medical University, Harbin 150081, China
- Department of Human Morphology, Okayama University Graduate School of Medicine, Density and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Atsunori Shitamukai
- Department of Human Morphology, Okayama University Graduate School of Medicine, Density and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Shucai Yang
- Human Anatomy and Histology and Embryology, School of Basic Medicine, Harbin Medical University, Harbin 150081, China
| | - Ayano Kawaguchi
- Department of Human Morphology, Okayama University Graduate School of Medicine, Density and Pharmaceutical Sciences, Okayama 700-8558, Japan
| |
Collapse
|
13
|
Sautreuil C, Lecointre M, Derambure C, Brasse-Lagnel C, Leroux P, Laquerrière A, Nicolas G, Gil S, Savage DD, Marret S, Marguet F, Falluel-Morel A, Gonzalez BJ. Prenatal Alcohol Exposure Impairs the Placenta-Cortex Transcriptomic Signature, Leading to Dysregulation of Angiogenic Pathways. Int J Mol Sci 2023; 24:13484. [PMID: 37686296 PMCID: PMC10488081 DOI: 10.3390/ijms241713484] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 09/10/2023] Open
Abstract
Although alcohol consumption during pregnancy is a major cause of behavioral and learning disabilities, most FASD infants are late- or even misdiagnosed due to clinician's difficulties achieving early detection of alcohol-induced neurodevelopmental impairments. Neuroplacentology has emerged as a new field of research focusing on the role of the placenta in fetal brain development. Several studies have reported that prenatal alcohol exposure (PAE) dysregulates a functional placenta-cortex axis, which is involved in the control of angiogenesis and leads to neurovascular-related defects. However, these studies were focused on PlGF, a pro-angiogenic factor. The aim of the present study is to provide the first transcriptomic "placenta-cortex" signature of the effects of PAE on fetal angiogenesis. Whole mouse genome microarrays of paired placentas and cortices were performed to establish the transcriptomic inter-organ "placenta-cortex" signature in control and PAE groups at gestational day 20. Genespring comparison of the control and PAE signatures revealed that 895 and 1501 genes were only detected in one of two placenta-cortex expression profiles, respectively. Gene ontology analysis indicated that 107 of these genes were associated with vascular development, and String protein-protein interaction analysis showed that they were associated with three functional clusters. PANTHER functional classification analysis indicated that "intercellular communication" was a significantly enriched biological process, and 27 genes were encoded for neuroactive ligand/receptors interactors. Protein validation experiments involving Western blot for one ligand-receptor couple (Agt/AGTR1/2) confirmed the transcriptomic data, and Pearson statistical analysis of paired placentas and fetal cortices revealed a negative correlation between placental Atg and cortical AGTR1, which was significantly impacted by PAE. In humans, a comparison of a 38WG control placenta with a 36WG alcohol-exposed placenta revealed low Agt immunolabeling in the syncytiotrophoblast layer of the alcohol case. In conclusion, this study establishes the first transcriptomic placenta-cortex signature of a developing mouse. The data show that PAE markedly unbalances this inter-organ signature; in particular, several ligands and/or receptors involved in the control of angiogenesis. These data support that PAE modifies the existing communication between the two organs and opens new research avenues regarding the impact of placental dysfunction on the neurovascular development of fetuses. Such a signature would present a clinical value for early diagnosis of brain defects in FASD.
Collapse
Affiliation(s)
- Camille Sautreuil
- University Rouen Normandie, INSERM U1245, Team Epigenetics and Pathophysiology of Neurodevelopmental Disorders, 76183 Rouen, France; (C.S.); (M.L.); (C.B.-L.); (P.L.); (A.L.); (S.M.); (F.M.); (A.F.-M.)
| | - Maryline Lecointre
- University Rouen Normandie, INSERM U1245, Team Epigenetics and Pathophysiology of Neurodevelopmental Disorders, 76183 Rouen, France; (C.S.); (M.L.); (C.B.-L.); (P.L.); (A.L.); (S.M.); (F.M.); (A.F.-M.)
| | - Céline Derambure
- University Rouen Normandie, INSERM U1245, Team Genetic Predisposition to Cancer, 76000 Rouen, France;
- Joint Genomics Facilities, Rouen University, 76183 Rouen, France
| | - Carole Brasse-Lagnel
- University Rouen Normandie, INSERM U1245, Team Epigenetics and Pathophysiology of Neurodevelopmental Disorders, 76183 Rouen, France; (C.S.); (M.L.); (C.B.-L.); (P.L.); (A.L.); (S.M.); (F.M.); (A.F.-M.)
| | - Philippe Leroux
- University Rouen Normandie, INSERM U1245, Team Epigenetics and Pathophysiology of Neurodevelopmental Disorders, 76183 Rouen, France; (C.S.); (M.L.); (C.B.-L.); (P.L.); (A.L.); (S.M.); (F.M.); (A.F.-M.)
| | - Annie Laquerrière
- University Rouen Normandie, INSERM U1245, Team Epigenetics and Pathophysiology of Neurodevelopmental Disorders, 76183 Rouen, France; (C.S.); (M.L.); (C.B.-L.); (P.L.); (A.L.); (S.M.); (F.M.); (A.F.-M.)
- Department of Pathology, Rouen University Hospital, 76183 Rouen, France
| | - Gaël Nicolas
- University Rouen Normandie, INSERM U1245, Team Genomics for Brain Disorders, 76183 Rouen, France;
| | - Sophie Gil
- INSERM UMR-S1144, Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France;
| | - Daniel D. Savage
- Department of Neurosciences, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA;
| | - Stéphane Marret
- University Rouen Normandie, INSERM U1245, Team Epigenetics and Pathophysiology of Neurodevelopmental Disorders, 76183 Rouen, France; (C.S.); (M.L.); (C.B.-L.); (P.L.); (A.L.); (S.M.); (F.M.); (A.F.-M.)
- Department of Neonatal Paediatrics and Intensive Care, Rouen University Hospital, University Rouen Normandie and CHU Rouen, 76183 Rouen, France
| | - Florent Marguet
- University Rouen Normandie, INSERM U1245, Team Epigenetics and Pathophysiology of Neurodevelopmental Disorders, 76183 Rouen, France; (C.S.); (M.L.); (C.B.-L.); (P.L.); (A.L.); (S.M.); (F.M.); (A.F.-M.)
- Department of Pathology, Rouen University Hospital, 76183 Rouen, France
| | - Anthony Falluel-Morel
- University Rouen Normandie, INSERM U1245, Team Epigenetics and Pathophysiology of Neurodevelopmental Disorders, 76183 Rouen, France; (C.S.); (M.L.); (C.B.-L.); (P.L.); (A.L.); (S.M.); (F.M.); (A.F.-M.)
| | - Bruno J. Gonzalez
- University Rouen Normandie, INSERM U1245, Team Epigenetics and Pathophysiology of Neurodevelopmental Disorders, 76183 Rouen, France; (C.S.); (M.L.); (C.B.-L.); (P.L.); (A.L.); (S.M.); (F.M.); (A.F.-M.)
| |
Collapse
|
14
|
Tabata H, Mori D, Matsuki T, Yoshizaki K, Asai M, Nakayama A, Ozaki N, Nagata KI. Histological Analysis of a Mouse Model of the 22q11.2 Microdeletion Syndrome. Biomolecules 2023; 13:biom13050763. [PMID: 37238632 DOI: 10.3390/biom13050763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
22q11.2 deletion syndrome (22q11.2DS) is associated with a high risk of developing various psychiatric and developmental disorders, including schizophrenia and early-onset Parkinson's disease. Recently, a mouse model of this disease, Del(3.0Mb)/+, mimicking the 3.0 Mb deletion which is most frequently found in patients with 22q11.2DS, was generated. The behavior of this mouse model was extensively studied and several abnormalities related to the symptoms of 22q11.2DS were found. However, the histological features of their brains have been little addressed. Here we describe the cytoarchitectures of the brains of Del(3.0Mb)/+ mice. First, we investigated the overall histology of the embryonic and adult cerebral cortices, but they were indistinguishable from the wild type. However, the morphologies of individual neurons were slightly but significantly changed from the wild type counterparts in a region-specific manner. The dendritic branches and/or dendritic spine densities of neurons in the medial prefrontal cortex, nucleus accumbens, and primary somatosensory cortex were reduced. We also observed reduced axon innervation of dopaminergic neurons into the prefrontal cortex. Given these affected neurons function together as the dopamine system to control animal behaviors, the impairment we observed may explain a part of the abnormal behaviors of Del(3.0Mb)/+ mice and the psychiatric symptoms of 22q11.2DS.
Collapse
Affiliation(s)
- Hidenori Tabata
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya, Kasugai 480-0392, Japan
| | - Daisuke Mori
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
- Brain and Mind Research Center, Nagoya University, Nagoya 466-8550, Japan
| | - Tohru Matsuki
- Department of Cellular Pathology, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya, Kasugai 480-0392, Japan
| | - Kaichi Yoshizaki
- Department of Disease Model, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya, Kasugai 480-0392, Japan
| | - Masato Asai
- Department of Disease Model, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya, Kasugai 480-0392, Japan
| | - Atsuo Nakayama
- Department of Cellular Pathology, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya, Kasugai 480-0392, Japan
- Department of Neurochemistry, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Norio Ozaki
- Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
- Institute for Glyco-core Research (iGCORE), Nagoya University, Chikusa-ku, Nagoya 464-0814, Japan
| | - Koh-Ichi Nagata
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, 713-8 Kamiya, Kasugai 480-0392, Japan
- Department of Neurochemistry, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|