1
|
Aquilino M, Ditzer N, Namba T, Albert M. Epigenetic and metabolic regulation of developmental timing in neocortex evolution. Trends Neurosci 2025:S0166-2236(25)00056-6. [PMID: 40155272 DOI: 10.1016/j.tins.2025.03.001] [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: 11/28/2024] [Revised: 02/13/2025] [Accepted: 03/03/2025] [Indexed: 04/01/2025]
Abstract
The human brain is characterized by impressive cognitive abilities. The neocortex is the seat of higher cognition, and neocortex expansion is a hallmark of human evolution. While developmental programs are similar in different species, the timing of developmental transitions and the capacity of neural progenitor cells (NPCs) to proliferate differ, contributing to the increased production of neurons during human cortical development. Here, we review the epigenetic regulation of developmental transitions during corticogenesis, focusing mostly on humans while building on knowledge from studies in mice. We discuss metabolic-epigenetic interplay as a potential mechanism to integrate extracellular signals into neural chromatin. Moreover, we synthesize current understanding of how epigenetic and metabolic deregulation can cause neurodevelopmental disorders. Finally, we outline how developmental timing can be investigated using brain organoid models.
Collapse
Affiliation(s)
- Matilde Aquilino
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Nora Ditzer
- Center for Regenerative Therapies Dresden, TUD Dresden University of Technology, 01307 Dresden, Germany
| | - Takashi Namba
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland; Department of Developmental Biology, Fujita Health University School of Medicine, Toyoake, Japan; International Center for Brain Science (ICBS), Fujita Health University, Toyoake, Japan.
| | - Mareike Albert
- Center for Regenerative Therapies Dresden, TUD Dresden University of Technology, 01307 Dresden, Germany.
| |
Collapse
|
2
|
Shimaoka K, Hori K, Miyashita S, Inoue YU, Tabe NKN, Sakamoto A, Hasegawa I, Nishitani K, Yamashiro K, Egusa SF, Tatsumoto S, Go Y, Abe M, Sakimura K, Inoue T, Imamura T, Hoshino M. The microcephaly-associated transcriptional regulator AUTS2 cooperates with Polycomb complex PRC2 to produce upper-layer neurons in mice. EMBO J 2025; 44:1354-1378. [PMID: 39815005 PMCID: PMC11876313 DOI: 10.1038/s44318-024-00343-7] [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: 04/04/2024] [Revised: 11/22/2024] [Accepted: 11/28/2024] [Indexed: 01/18/2025] Open
Abstract
AUTS2 syndrome is characterized by intellectual disability and microcephaly, and is often associated with autism spectrum disorder, but the underlying mechanisms, particularly concerning microcephaly, remain incompletely understood. Here, we analyze mice mutated for the transcriptional regulator AUTS2, which recapitulate microcephaly. Their brains exhibit reduced division of intermediate progenitor cells (IPCs), leading to fewer neurons and decreased thickness in the upper-layer cortex. Increased expression of the AUTS2 transcriptional target Robo1 in the mutant animals suppresses IPC division, and transcriptomic and chromatin profiling shows that AUTS2 primarily represses transcription of genes like Robo1 in IPCs. Regions around the transcriptional start sites of AUTS2 target genes are enriched for the repressive histone modification H3K27me3, which is reduced in Auts2 mutants. Furthermore, we find that AUTS2 interacts with Polycomb complex PRC2, with which it cooperates to promote IPC division. These findings shed light on the microcephaly phenotype observed in the AUTS2 syndrome.
Collapse
Affiliation(s)
- Kazumi Shimaoka
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, 187-8502, Japan
| | - Kei Hori
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, 187-8502, Japan
| | - Satoshi Miyashita
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, 187-8502, Japan
| | - Yukiko U Inoue
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, 187-8502, Japan
| | - Nao K N Tabe
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, 187-8502, Japan
- Department of NCNP Brain Physiology and Pathology, Institute of Science Tokyo, Tokyo, 113-8510, Japan
| | - Asami Sakamoto
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, 187-8502, Japan
| | - Ikuko Hasegawa
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, 187-8502, Japan
| | - Kayo Nishitani
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, 187-8502, Japan
| | - Kunihiko Yamashiro
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, 187-8502, Japan
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Saki F Egusa
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, 187-8502, Japan
| | - Shoji Tatsumoto
- Cognitive Genomics Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, 444-8585, Japan
| | - Yasuhiro Go
- Cognitive Genomics Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi, 444-8585, Japan
- Department of System Neuroscience, Division of Behavioral Development, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi, 444-8585, Japan
- Graduate School of Information Science, University of Hyogo, Kobe, Hyogo, 650-0047, Japan
| | - Manabu Abe
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Kenji Sakimura
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, 951-8585, Japan
| | - Takayoshi Inoue
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, 187-8502, Japan
| | - Takuya Imamura
- Program of Biomedical Science, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, 739-8526, Japan
| | - Mikio Hoshino
- Department of Biochemistry and Cellular Biology, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, 187-8502, Japan.
- Department of NCNP Brain Physiology and Pathology, Institute of Science Tokyo, Tokyo, 113-8510, Japan.
| |
Collapse
|
3
|
Pelenyi A, Atterton C, Jones J, Currey L, Al-Khalily M, Wright L, Kurniawan ND, Thor S, Piper M. Expression of the Hippo pathway effector, TEAD1, within the developing murine forebrain. Gene Expr Patterns 2024; 54:119384. [PMID: 39557142 DOI: 10.1016/j.gep.2024.119384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/06/2024] [Accepted: 11/15/2024] [Indexed: 11/20/2024]
Abstract
The Hippo pathway is a critical regulator of animal development. Activation of the Hippo pathway causes a cascade of phosphorylation events that culminate in the phosphorylation of the transcriptional co-factors YAP and TAZ, which limits their entry into the nucleus. When the Hippo pathway is 'off', however, YAP and TAZ can enter the nucleus, where they interact with the transcription factors of the TEA Domain (TEAD) family to regulate transcriptional activity. Despite the importance of the Hippo pathway for development, including within the nervous system, the expression of the TEAD family remains poorly defined in mammals. Here, we mapped the expression of TEAD1 in the developing mouse brain. We find that TEAD1 expression is confined to progenitor cells during embryonic development, namely radial glia and intermediate progenitor cells. TEAD1 expression is not evident in post-mitotic neurons of the cortical plate. We also identify expression of TEAD1 in developing and mature ependymal cells of the lateral and third ventricle, including within the subcommissural organ, as well as by cells within the choroid plexuses and the forebrain neurogenic niches. Finally, we find that adult mice conditionally heterozygous for Tead1 in the central nervous system exhibit a significantly smaller brain. Collectively, these findings reveal a specific pattern of expression for TEAD1 during telencephalic development and implicate this factor in regulating neural progenitor cell proliferation.
Collapse
Affiliation(s)
- Alexandra Pelenyi
- The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD, 4072, Australia
| | - Cooper Atterton
- The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD, 4072, Australia
| | - Justin Jones
- The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD, 4072, Australia
| | - Laura Currey
- The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD, 4072, Australia
| | - Majd Al-Khalily
- The Centre for Advanced Imaging, The University of Queensland, QLD, 4072, Australia
| | - Lucinda Wright
- The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD, 4072, Australia
| | - Nyoman D Kurniawan
- The Centre for Advanced Imaging, The University of Queensland, QLD, 4072, Australia
| | - Stefan Thor
- The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD, 4072, Australia
| | - Michael Piper
- The School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, QLD, 4072, Australia; The Queensland Brain Institute, The University of Queensland, QLD, 4072, Australia.
| |
Collapse
|
4
|
Imamura M, Yoshino M, Kawasaki H. Investigation of the development and evolution of the mammalian cerebrum using gyrencephalic ferrets. Eur J Cell Biol 2024; 103:151466. [PMID: 39546916 DOI: 10.1016/j.ejcb.2024.151466] [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: 08/31/2024] [Revised: 11/02/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024] Open
Abstract
Mammalian brains have evolved a neocortex, which has diverged in size and morphology in different species over the course of evolution. In some mammals, a substantial increase in the number of neurons and glial cells resulted in the expansion and folding of the cerebrum, and it is believed that these evolutionary changes contributed to the acquisition of higher cognitive abilities in mammals. However, their underlying molecular and cellular mechanisms remain insufficiently elucidated. A major difficulty in addressing these mechanisms stemmed from the lack of appropriate animal models, as conventional experimental animals such as mice and rats have small brains without structurally obvious folds. Therefore, researchers including us have focused on using ferrets instead of mice and rats. Ferrets are domesticated carnivorous mammals with a gyrencephalic cerebrum, and, notably, they are amenable to genetic manipulations including in utero electroporation to knock out genes in the cerebrum. In this review, we highlight recent research into the mechanisms underlying the development and evolution of cortical folds using ferrets.
Collapse
Affiliation(s)
- Masanori Imamura
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan; Sapiens Life Sciences, Evolution and Medicine Research Center, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| | - Mayuko Yoshino
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan; Sapiens Life Sciences, Evolution and Medicine Research Center, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan
| | - Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan; Sapiens Life Sciences, Evolution and Medicine Research Center, Kanazawa University, Kanazawa, Ishikawa 920-8640, Japan.
| |
Collapse
|
5
|
Nakamura Y, Shimada IS, Maroofian R, Falabella M, Zaki MS, Fujimoto M, Sato E, Takase H, Aoki S, Miyauchi A, Koshimizu E, Miyatake S, Arioka Y, Honda M, Higashi T, Miya F, Okubo Y, Ogawa I, Scardamaglia A, Miryounesi M, Alijanpour S, Ahmadabadi F, Herkenrath P, Dafsari HS, Velmans C, Al Balwi M, Vitobello A, Denommé-Pichon AS, Jeanne M, Civit A, Abdel-Hamid MS, Naderi H, Darvish H, Bakhtiari S, Kruer MC, Carroll CJ, Ghayoor Karimiani E, Khailany RA, Abdulqadir TA, Ozaslan M, Bauer P, Zifarelli G, Seifi T, Zamani M, Al Alam C, Alvi JR, Sultan T, Efthymiou S, Pope SAS, Haginoya K, Matsunaga T, Osaka H, Matsumoto N, Ozaki N, Ohkawa Y, Oki S, Tsunoda T, Pitceathly RDS, Taketomi Y, Houlden H, Murakami M, Kato Y, Saitoh S. Biallelic null variants in PNPLA8 cause microcephaly by reducing the number of basal radial glia. Brain 2024; 147:3949-3967. [PMID: 39082157 PMCID: PMC11531855 DOI: 10.1093/brain/awae185] [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: 05/18/2023] [Revised: 05/05/2024] [Accepted: 05/20/2024] [Indexed: 11/05/2024] Open
Abstract
Patatin-like phospholipase domain-containing lipase 8 (PNPLA8), one of the calcium-independent phospholipase A2 enzymes, is involved in various physiological processes through the maintenance of membrane phospholipids. Biallelic variants in PNPLA8 have been associated with a range of paediatric neurodegenerative disorders. However, the phenotypic spectrum, genotype-phenotype correlations and the underlying mechanisms are poorly understood. Here, we newly identified 14 individuals from 12 unrelated families with biallelic ultra-rare variants in PNPLA8 presenting with a wide phenotypic spectrum of clinical features. Analysis of the clinical features of current and previously reported individuals (25 affected individuals across 20 families) showed that PNPLA8-related neurological diseases manifest as a continuum ranging from variable developmental and/or degenerative epileptic-dyskinetic encephalopathy to childhood-onset neurodegeneration. We found that complete loss of PNPLA8 was associated with the more profound end of the spectrum, with congenital microcephaly. Using cerebral organoids generated from human induced pluripotent stem cells, we found that loss of PNPLA8 led to developmental defects by reducing the number of basal radial glial cells and upper-layer neurons. Spatial transcriptomics revealed that loss of PNPLA8 altered the fate specification of apical radial glial cells, as reflected by the enrichment of gene sets related to the cell cycle, basal radial glial cells and neural differentiation. Neural progenitor cells lacking PNPLA8 showed a reduced amount of lysophosphatidic acid, lysophosphatidylethanolamine and phosphatidic acid. The reduced number of basal radial glial cells in patient-derived cerebral organoids was rescued, in part, by the addition of lysophosphatidic acid. Our data suggest that PNPLA8 is crucial to meet phospholipid synthetic needs and to produce abundant basal radial glial cells in human brain development.
Collapse
Affiliation(s)
- Yuji Nakamura
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| | - Issei S Shimada
- Department of Cell Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| | - Reza Maroofian
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Micol Falabella
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Maha S Zaki
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo 12622, Egypt
| | - Masanori Fujimoto
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| | - Emi Sato
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| | - Hiroshi Takase
- Core Laboratory, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| | - Shiho Aoki
- Department of Pediatrics, Jichi Medical University, Tochigi 3290498, Japan
| | - Akihiko Miyauchi
- Department of Pediatrics, Jichi Medical University, Tochigi 3290498, Japan
| | - Eriko Koshimizu
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama 2360004, Japan
| | - Satoko Miyatake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama 2360004, Japan
- Department of Clinical Genetics, Yokohama City University Hospital, Yokohama 2360004, Japan
| | - Yuko Arioka
- Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya 4668550, Japan
| | - Mizuki Honda
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
- Laboratory of Molecular and Cellular Physiology, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima 7398526, Japan
| | - Takayoshi Higashi
- Laboratory of Microenvironmental and Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 1138655, Japan
| | - Fuyuki Miya
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, 1608582, Japan
| | - Yukimune Okubo
- Department of Pediatric Neurology, Miyagi Children's Hospital, Sendai 9893126, Japan
| | - Isamu Ogawa
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 4678603, Japan
| | - Annarita Scardamaglia
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Mohammad Miryounesi
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1516745811, Iran
| | - Sahar Alijanpour
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1516745811, Iran
| | - Farzad Ahmadabadi
- Pediatric Neurology Department, Faculty of Medicine, Mofid Children's Hospital, Shahid Beheshti University of Medical Sciences, Tehran 1546815514, Iran
| | - Peter Herkenrath
- Department of Pediatrics and Center for Rare Diseases, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne 50937, Germany
| | - Hormos Salimi Dafsari
- Department of Pediatrics and Center for Rare Diseases, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne 50937, Germany
- Max-Planck-Institute for Biology of Ageing, Cologne 50931, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne 50931, Germany
| | - Clara Velmans
- Faculty of Medicine and University Hospital Cologne, Institute of Human Genetics, University of Cologne, Cologne 50931, Germany
| | - Mohammed Al Balwi
- Department of Pathology and Laboratory Medicine, College of Medicine, KSAU-HS, Ministry of National Guard Health Affairs, Riyadh 11426, Saudi Arabia
| | - Antonio Vitobello
- Functional Unit for Diagnostic Innovation in Rare Diseases, FHU-TRANSLAD, Dijon Bourgogne University Hospital, Dijon 21000, France
- INSERM UMR1231 GAD ‘Génétique des Anomalies du Développement’, FHU-TRANSLAD, University of Burgundy, Dijon 21000, France
| | - Anne-Sophie Denommé-Pichon
- Functional Unit for Diagnostic Innovation in Rare Diseases, FHU-TRANSLAD, Dijon Bourgogne University Hospital, Dijon 21000, France
- INSERM UMR1231 GAD ‘Génétique des Anomalies du Développement’, FHU-TRANSLAD, University of Burgundy, Dijon 21000, France
| | - Médéric Jeanne
- Genetics Department, University Hospital of Tours, Tours 37044, France
- UMR 1253, iBrain, University of Tours, INSERM, Tours 37032, France
| | - Antoine Civit
- Genetics Department, University Hospital of Tours, Tours 37044, France
| | - Mohamed S Abdel-Hamid
- Medical Molecular Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo 12622, Egypt
| | - Hamed Naderi
- Neuroscience Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan 4918936316, Iran
| | - Hossein Darvish
- Neuroscience Research Center, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan 4918936316, Iran
| | - Somayeh Bakhtiari
- Pediatric Movement Disorders Program, Division of Pediatric Neurology, Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, AZ 85016, USA
- Departments of Child Health, Neurology, Cellular & Molecular Medicine and Program in Genetics, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Michael C Kruer
- Pediatric Movement Disorders Program, Division of Pediatric Neurology, Barrow Neurological Institute, Phoenix Children's Hospital, Phoenix, AZ 85016, USA
- Departments of Child Health, Neurology, Cellular & Molecular Medicine and Program in Genetics, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Christopher J Carroll
- Genetics Section, Molecular and Clinical Sciences Research Institute, St. George's, University of London, London SW17 0RE, UK
| | - Ehsan Ghayoor Karimiani
- Genetics Section, Molecular and Clinical Sciences Research Institute, St. George's, University of London, London SW17 0RE, UK
| | - Rozhgar A Khailany
- Department of Basic Science, Hawler Medical University, Erbil, Kurdistan Region 44001, Iraq
| | - Talib Adil Abdulqadir
- Department of Pediatrics, College of Medicine, Hawler Medical University, Erbil, Kurdistan Region 44001, Iraq
| | - Mehmet Ozaslan
- Department of Biology, Division of Molecular Biology and Genetics, Gaziantep University, Gaziantep 27410, Turkey
| | | | | | - Tahere Seifi
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz 83151-61355, Iran
- Narges Medical Genetics and Prenatal Diagnosis Laboratory, Kianpars, Ahvaz 61556-89467, Iran
| | - Mina Zamani
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz 83151-61355, Iran
- Narges Medical Genetics and Prenatal Diagnosis Laboratory, Kianpars, Ahvaz 61556-89467, Iran
| | - Chadi Al Alam
- Pediatrics and Pediatric Neurology, American Center for Psychiatry and Neurology, Abu Dhabi 108699, UAE
| | - Javeria Raza Alvi
- Department of Pediatric Neurology, the Children’s Hospital and the University of Child Health Sciences, Lahore 54600, Pakistan
| | - Tipu Sultan
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Stephanie Efthymiou
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Simon A S Pope
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
- Neurometabolic Unit, The National Hospital for Neurology and Neurosurgery, London WC1N 3BG, UK
| | - Kazuhiro Haginoya
- Department of Pediatric Neurology, Miyagi Children's Hospital, Sendai 9893126, Japan
| | - Tamihide Matsunaga
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 4678603, Japan
| | - Hitoshi Osaka
- Department of Pediatrics, Jichi Medical University, Tochigi 3290498, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama 2360004, Japan
| | - Norio Ozaki
- Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, Nagoya 4668550, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 8128582, Japan
| | - Shinya Oki
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto 6068507, Japan
- Institute of Resource Development and Analysis, Kumamoto University, Kumamoto 8600811, Japan
| | - Tatsuhiko Tsunoda
- Laboratory for Medical Science Mathematics, Department of Biological Sciences, School of Science, The University of Tokyo, Tokyo 113-0033, Japan
- Laboratory for Medical Science Mathematics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo 113-0033, Japan
- Laboratory for Medical Science Mathematics, RIKEN Center for Integrative Medical Sciences, Yokohama 230-0045, Japan
| | - Robert D S Pitceathly
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London WC1N 3BG, UK
| | - Yoshitaka Taketomi
- Laboratory of Microenvironmental and Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 1138655, Japan
| | - Henry Houlden
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Makoto Murakami
- Laboratory of Microenvironmental and Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 1138655, Japan
| | - Yoichi Kato
- Department of Cell Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| | - Shinji Saitoh
- Department of Pediatrics and Neonatology, Nagoya City University Graduate School of Medical Sciences, Nagoya 4678601, Japan
| |
Collapse
|
6
|
Thor S. Indirect neurogenesis in space and time. Nat Rev Neurosci 2024; 25:519-534. [PMID: 38951687 DOI: 10.1038/s41583-024-00833-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2024] [Indexed: 07/03/2024]
Abstract
During central nervous system (CNS) development, neural progenitor cells (NPCs) generate neurons and glia in two different ways. In direct neurogenesis, daughter cells differentiate directly into neurons or glia, whereas in indirect neurogenesis, neurons or glia are generated after one or more daughter cell divisions. Intriguingly, indirect neurogenesis is not stochastically deployed and plays instructive roles during CNS development: increased generation of cells from specific lineages; increased generation of early or late-born cell types within a lineage; and increased cell diversification. Increased indirect neurogenesis might contribute to the anterior CNS expansion evident throughout the Bilateria and help to modify brain-region size without requiring increased NPC numbers or extended neurogenesis. Increased indirect neurogenesis could be an evolutionary driver of the gyrencephalic (that is, folded) cortex that emerged during mammalian evolution and might even have increased during hominid evolution. Thus, selection of indirect versus direct neurogenesis provides a powerful developmental and evolutionary instrument that drives not only the evolution of CNS complexity but also brain expansion and modulation of brain-region size, and thereby the evolution of increasingly advanced cognitive abilities. This Review describes indirect neurogenesis in several model species and humans, and highlights some of the molecular genetic mechanisms that control this important process.
Collapse
Affiliation(s)
- Stefan Thor
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia.
| |
Collapse
|
7
|
Chen J, Tsai YH, Linden AK, Kessler JA, Peng CY. YAP and TAZ differentially regulate postnatal cortical progenitor proliferation and astrocyte differentiation. J Cell Sci 2024; 137:jcs261516. [PMID: 38639242 DOI: 10.1242/jcs.261516] [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: 07/25/2023] [Accepted: 04/09/2024] [Indexed: 04/20/2024] Open
Abstract
WW domain-containing transcription regulator 1 (WWTR1, referred to here as TAZ) and Yes-associated protein (YAP, also known as YAP1) are transcriptional co-activators traditionally studied together as a part of the Hippo pathway, and are best known for their roles in stem cell proliferation and differentiation. Despite their similarities, TAZ and YAP can exert divergent cellular effects by differentially interacting with other signaling pathways that regulate stem cell maintenance or differentiation. In this study, we show in mouse neural stem and progenitor cells (NPCs) that TAZ regulates astrocytic differentiation and maturation, and that TAZ mediates some, but not all, of the effects of bone morphogenetic protein (BMP) signaling on astrocytic development. By contrast, both TAZ and YAP mediate the effects on NPC fate of β1-integrin (ITGB1) and integrin-linked kinase signaling, and these effects are dependent on extracellular matrix cues. These findings demonstrate that TAZ and YAP perform divergent functions in the regulation of astrocyte differentiation, where YAP regulates cell cycle states of astrocytic progenitors and TAZ regulates differentiation and maturation from astrocytic progenitors into astrocytes.
Collapse
Affiliation(s)
- Jessie Chen
- Department of Neurology, Northwestern University's Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yung-Hsu Tsai
- Department of Neurology, Northwestern University's Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Anne K Linden
- Department of Neurology, Northwestern University's Feinberg School of Medicine, Chicago, IL 60611, USA
| | - John A Kessler
- Department of Neurology, Northwestern University's Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Chian-Yu Peng
- Department of Neurology, Northwestern University's Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
8
|
Kawasaki H. Investigation of the mechanisms underlying the development and evolution of folds of the cerebrum using gyrencephalic ferrets. J Comp Neurol 2024; 532:e25615. [PMID: 38587214 DOI: 10.1002/cne.25615] [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: 12/24/2023] [Revised: 02/22/2024] [Accepted: 03/24/2024] [Indexed: 04/09/2024]
Abstract
The mammalian cerebrum has changed substantially during evolution, characterized by increases in neurons and glial cells and by the expansion and folding of the cerebrum. While these evolutionary alterations are thought to be crucial for acquiring higher cognitive functions, the molecular mechanisms underlying the development and evolution of the mammalian cerebrum remain only partially understood. This is, in part, because of the difficulty in analyzing these mechanisms using mice only. To overcome this limitation, genetic manipulation techniques for the cerebrum of gyrencephalic carnivore ferrets have been developed. Furthermore, successful gene knockout in the ferret cerebrum has been accomplished through the application of the CRISPR/Cas9 system. This review mainly highlights recent research conducted using gyrencephalic carnivore ferrets to investigate the mechanisms underlying the development and evolution of cortical folds.
Collapse
Affiliation(s)
- Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
9
|
Li MY, Yang XL, Chung CC, Lai YJ, Tsai JC, Kuo YL, Yu JY, Wang TW. TRIP6 promotes neural stem cell maintenance through YAP-mediated Sonic Hedgehog activation. FASEB J 2024; 38:e23501. [PMID: 38411462 DOI: 10.1096/fj.202301805rrr] [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: 09/13/2023] [Revised: 01/24/2024] [Accepted: 02/06/2024] [Indexed: 02/28/2024]
Abstract
In the adult mammalian brain, new neurons are continuously generated from neural stem cells (NSCs) in the subventricular zone (SVZ)-olfactory bulb (OB) pathway. YAP, a transcriptional co-activator of the Hippo pathway, promotes cell proliferation and inhibits differentiation in embryonic neural progenitors. However, the role of YAP in postnatal NSCs remains unclear. Here, we showed that YAP was present in NSCs of the postnatal mouse SVZ. Forced expression of Yap promoted NSC maintenance and inhibited differentiation, whereas depletion of Yap by RNA interference or conditional knockout led to the decline of NSC maintenance, premature neuronal differentiation, and collapse of neurogenesis. For the molecular mechanism, thyroid hormone receptor-interacting protein 6 (TRIP6) recruited protein phosphatase PP1A to dephosphorylate LATS1/2, therefore inducing YAP nuclear localization and activation. Moreover, TRIP6 promoted NSC maintenance, cell proliferation, and inhibited differentiation through YAP. In addition, YAP regulated the expression of the Sonic Hedgehog (SHH) pathway effector Gli2 and Gli1/2 mediated the effect of YAP on NSC maintenance. Together, our findings demonstrate a novel TRIP6-YAP-SHH axis, which is critical for regulating postnatal neurogenesis in the SVZ-OB pathway.
Collapse
Affiliation(s)
- Ming-Yang Li
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Xiu-Li Yang
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Chia-Chi Chung
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Yun-Ju Lai
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Jui-Cheng Tsai
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Ya-Lin Kuo
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Jenn-Yah Yu
- Department of Life Sciences, Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tsu-Wei Wang
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| |
Collapse
|
10
|
Fan W, Jurado‐Arjona J, Alanis‐Lobato G, Péron S, Berger C, Andrade‐Navarro MA, Falk S, Berninger B. The transcriptional co-activator Yap1 promotes adult hippocampal neural stem cell activation. EMBO J 2023; 42:e110384. [PMID: 37083045 PMCID: PMC10233373 DOI: 10.15252/embj.2021110384] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/20/2023] [Accepted: 03/27/2023] [Indexed: 04/22/2023] Open
Abstract
Most adult hippocampal neural stem cells (NSCs) remain quiescent, with only a minor portion undergoing active proliferation and neurogenesis. The molecular mechanisms that trigger the transition from quiescence to activation are still poorly understood. Here, we found the activity of the transcriptional co-activator Yap1 to be enriched in active NSCs. Genetic deletion of Yap1 led to a significant reduction in the relative proportion of active NSCs, supporting a physiological role of Yap1 in regulating the transition from quiescence to activation. Overexpression of wild-type Yap1 in adult NSCs did not induce NSC activation, suggesting tight upstream control mechanisms, but overexpression of a gain-of-function mutant (Yap1-5SA) elicited cell cycle entry in NSCs and hilar astrocytes. Consistent with a role of Yap1 in NSC activation, single cell RNA sequencing revealed a partial induction of an activated NSC gene expression program. Furthermore, Yap1-5SA expression also induced expression of Taz and other key components of the Yap/Taz regulon that were previously identified in glioblastoma stem cell-like cells. Consequently, dysregulated Yap1 activity led to repression of hippocampal neurogenesis, aberrant cell differentiation, and partial acquisition of a glioblastoma stem cell-like signature.
Collapse
Affiliation(s)
- Wenqiang Fan
- Institute of Physiological ChemistryUniversity Medical Center of the Johannes Gutenberg University MainzMainzGermany
- Present address:
Neuroscience Therapeutic Area, New MedicinesUCB Biopharma SPRLBraine‐l'AlleudBelgium
| | - Jerónimo Jurado‐Arjona
- Institute of Physiological ChemistryUniversity Medical Center of the Johannes Gutenberg University MainzMainzGermany
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK
| | - Gregorio Alanis‐Lobato
- Faculty of BiologyJohannes Gutenberg University MainzMainzGermany
- Present address:
Global Computational Biology and Data SciencesBoehringer Ingelheim Pharma GmbH & Co. KGBiberach an der RissGermany
| | - Sophie Péron
- Institute of Physiological ChemistryUniversity Medical Center of the Johannes Gutenberg University MainzMainzGermany
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK
| | - Christian Berger
- Institute of GeneticsJohannes Gutenberg University MainzMainzGermany
| | | | - Sven Falk
- Institute of BiochemistryFriedrich‐Alexander‐Universität Nürnberg‐ErlangenErlangenGermany
| | - Benedikt Berninger
- Institute of Physiological ChemistryUniversity Medical Center of the Johannes Gutenberg University MainzMainzGermany
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK
- The Francis Crick InstituteLondonUK
- Focus Program Translational NeuroscienceJohannes Gutenberg University MainzMainzGermany
| |
Collapse
|
11
|
Fernández V, Borrell V. Developmental mechanisms of gyrification. Curr Opin Neurobiol 2023; 80:102711. [DOI: 10.1016/j.conb.2023.102711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 02/09/2023] [Accepted: 02/23/2023] [Indexed: 03/31/2023]
|
12
|
Gkini V, Namba T. Glutaminolysis and the Control of Neural Progenitors in Neocortical Development and Evolution. Neuroscientist 2023; 29:177-189. [PMID: 35057642 PMCID: PMC10018057 DOI: 10.1177/10738584211069060] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Multiple types of neural progenitor cells (NPCs) contribute to the development of the neocortex, a brain region responsible for our higher cognitive abilities. Proliferative capacity of NPCs varies among NPC types, developmental stages, and species. The higher proliferative capacity of NPCs in the developing human neocortex is thought to be a major contributing factor why humans have the most expanded neocortex within primates. Recent studies have shed light on the importance of cell metabolism in the neocortical NPC proliferative capacity. Specifically, glutaminolysis, a metabolic pathway that converts glutamine to glutamate and then to α-ketoglutarate, has been shown to play a critical role in human NPCs, both in apical and basal progenitors. In this review, we summarize our current knowledge of NPC metabolism, focusing especially on glutaminolysis, and discuss the role of NPC metabolism in neocortical development, evolution, and neurodevelopmental disorders, providing a broader perspective on a newly emerging research field.
Collapse
Affiliation(s)
- Vasiliki Gkini
- Neuroscience Center, HiLIFE—Helsinki
Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Takashi Namba
- Neuroscience Center, HiLIFE—Helsinki
Institute of Life Science, University of Helsinki, Helsinki, Finland
- Takashi Namba, Neuroscience Center, HiLIFE
— Helsinki Institute of Life Science, University of Helsinki, PO 63,
Haartmaninkatu 8, Helsinki 00014, Finland.
| |
Collapse
|
13
|
Van Heurck R, Bonnefont J, Wojno M, Suzuki IK, Velez-Bravo FD, Erkol E, Nguyen DT, Herpoel A, Bilheu A, Beckers S, Ledent C, Vanderhaeghen P. CROCCP2 acts as a human-specific modifier of cilia dynamics and mTOR signaling to promote expansion of cortical progenitors. Neuron 2023; 111:65-80.e6. [PMID: 36334595 PMCID: PMC9831670 DOI: 10.1016/j.neuron.2022.10.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/12/2022] [Accepted: 10/09/2022] [Indexed: 11/06/2022]
Abstract
The primary cilium is a central signaling component during embryonic development. Here we focus on CROCCP2, a hominid-specific gene duplicate from ciliary rootlet coiled coil (CROCC), also known as rootletin, that encodes the major component of the ciliary rootlet. We find that CROCCP2 is highly expressed in the human fetal brain and not in other primate species. CROCCP2 gain of function in the mouse embryonic cortex and human cortical cells and organoids results in decreased ciliogenesis and increased cortical progenitor amplification, particularly basal progenitors. CROCCP2 decreases ciliary dynamics by inhibition of the IFT20 ciliary trafficking protein, which then impacts neurogenesis through increased mTOR signaling. Loss of function of CROCCP2 in human cortical cells and organoids leads to increased ciliogenesis, decreased mTOR signaling, and impaired basal progenitor amplification. These data identify CROCCP2 as a human-specific modifier of cortical neurogenesis that acts through modulation of ciliary dynamics and mTOR signaling.
Collapse
Affiliation(s)
- Roxane Van Heurck
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium,Department of Neurosciences, Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium,Université Libre de Bruxelles (U.L.B.), Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM), and ULB Neuroscience Institute (UNI), 1070 Brussels, Belgium
| | - Jérôme Bonnefont
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium,Department of Neurosciences, Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium,Université Libre de Bruxelles (U.L.B.), Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM), and ULB Neuroscience Institute (UNI), 1070 Brussels, Belgium
| | - Marta Wojno
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium,Department of Neurosciences, Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium,Université Libre de Bruxelles (U.L.B.), Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM), and ULB Neuroscience Institute (UNI), 1070 Brussels, Belgium
| | - Ikuo K. Suzuki
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium,Department of Neurosciences, Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium,Université Libre de Bruxelles (U.L.B.), Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM), and ULB Neuroscience Institute (UNI), 1070 Brussels, Belgium,Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Fausto D. Velez-Bravo
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium,Department of Neurosciences, Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium,Université Libre de Bruxelles (U.L.B.), Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM), and ULB Neuroscience Institute (UNI), 1070 Brussels, Belgium
| | - Emir Erkol
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium,Department of Neurosciences, Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium,Université Libre de Bruxelles (U.L.B.), Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM), and ULB Neuroscience Institute (UNI), 1070 Brussels, Belgium
| | - Dan Truc Nguyen
- Université Libre de Bruxelles (U.L.B.), Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM), and ULB Neuroscience Institute (UNI), 1070 Brussels, Belgium
| | - Adèle Herpoel
- Université Libre de Bruxelles (U.L.B.), Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM), and ULB Neuroscience Institute (UNI), 1070 Brussels, Belgium
| | - Angéline Bilheu
- Université Libre de Bruxelles (U.L.B.), Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM), and ULB Neuroscience Institute (UNI), 1070 Brussels, Belgium
| | - Sofie Beckers
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium,Department of Neurosciences, Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium
| | - Catherine Ledent
- Université Libre de Bruxelles (U.L.B.), Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM), and ULB Neuroscience Institute (UNI), 1070 Brussels, Belgium
| | - Pierre Vanderhaeghen
- VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium,Department of Neurosciences, Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium,Université Libre de Bruxelles (U.L.B.), Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM), and ULB Neuroscience Institute (UNI), 1070 Brussels, Belgium,Corresponding author
| |
Collapse
|
14
|
Terry BK, Kim S. The Role of Hippo-YAP/TAZ Signaling in Brain Development. Dev Dyn 2022; 251:1644-1665. [PMID: 35651313 DOI: 10.1002/dvdy.504] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 05/19/2022] [Accepted: 05/24/2022] [Indexed: 11/08/2022] Open
Abstract
In order for our complex nervous system to develop normally, both precise spatial and temporal regulation of a number of different signaling pathways is critical. During both early embryogenesis and in organ development, one pathway that has been repeatedly implicated is the Hippo-YAP/TAZ signaling pathway. The paralogs YAP and TAZ are transcriptional co-activators that play an important role in cell proliferation, cell differentiation, and organ growth. Regulation of these proteins by the Hippo kinase cascade is therefore important for normal development. In this article, we review the growing field of research surrounding the role of Hippo-YAP/TAZ signaling in normal and atypical brain development. Starting from the development of the neural tube to the development and refinement of the cerebral cortex, cerebellum, and ventricular system, we address the typical role of these transcriptional co-activators, the functional consequences that manipulation of YAP/TAZ and their upstream regulators have on brain development, and where further research may be of benefit. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Bethany K Terry
- Shriners Hospitals Pediatrics Research Center, Department of Neural Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA.,Biomedical Sciences Graduate Program, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Seonhee Kim
- Shriners Hospitals Pediatrics Research Center, Department of Neural Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA
| |
Collapse
|
15
|
Casas Gimeno G, Paridaen JTML. The Symmetry of Neural Stem Cell and Progenitor Divisions in the Vertebrate Brain. Front Cell Dev Biol 2022; 10:885269. [PMID: 35693936 PMCID: PMC9174586 DOI: 10.3389/fcell.2022.885269] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/20/2022] [Indexed: 12/23/2022] Open
Abstract
Robust brain development requires the tight coordination between tissue growth, neuronal differentiation and stem cell maintenance. To achieve this, neural stem cells need to balance symmetric proliferative and terminal divisions with asymmetric divisions. In recent years, the unequal distribution of certain cellular components in mitosis has emerged as a key mechanism to regulate the symmetry of division, and the determination of equal and unequal sister cell fates. Examples of such components include polarity proteins, signaling components, and cellular structures such as endosomes and centrosomes. In several types of neural stem cells, these factors show specific patterns of inheritance that correlate to specific cell fates, albeit the underlying mechanism and the potential causal relationship is not always understood. Here, we review these examples of cellular neural stem and progenitor cell asymmetries and will discuss how they fit into our current understanding of neural stem cell function in neurogenesis in developing and adult brains. We will focus mainly on the vertebrate brain, though we will incorporate relevant examples from invertebrate organisms as well. In particular, we will highlight recent advances in our understanding of the complexities related cellular asymmetries in determining division mode outcomes, and how these mechanisms are spatiotemporally regulated to match the different needs for proliferation and differentiation as the brain forms.
Collapse
|
16
|
Jabali A, Hoffrichter A, Uzquiano A, Marsoner F, Wilkens R, Siekmann M, Bohl B, Rossetti AC, Horschitz S, Koch P, Francis F, Ladewig J. Human cerebral organoids reveal progenitor pathology in EML1-linked cortical malformation. EMBO Rep 2022; 23:e54027. [PMID: 35289477 PMCID: PMC9066063 DOI: 10.15252/embr.202154027] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 02/11/2022] [Accepted: 02/17/2022] [Indexed: 12/26/2022] Open
Abstract
Malformations of human cortical development (MCD) can cause severe disabilities. The lack of human‐specific models hampers our understanding of the molecular underpinnings of the intricate processes leading to MCD. Here, we use cerebral organoids derived from patients and genome edited‐induced pluripotent stem cells to address pathophysiological changes associated with a complex MCD caused by mutations in the echinoderm microtubule‐associated protein‐like 1 (EML1) gene. EML1‐deficient organoids display ectopic neural rosettes at the basal side of the ventricular zone areas and clusters of heterotopic neurons. Single‐cell RNA sequencing shows an upregulation of basal radial glial (RG) markers and human‐specific extracellular matrix components in the ectopic cell population. Gene ontology and molecular analyses suggest that ectopic progenitor cells originate from perturbed apical RG cell behavior and yes‐associated protein 1 (YAP1)‐triggered expansion. Our data highlight a progenitor origin of EML1 mutation‐induced MCD and provide new mechanistic insight into the human disease pathology.
Collapse
Affiliation(s)
- Ammar Jabali
- Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany.,Hector Institute for Translational Brain Research, Mannheim, Germany.,German Cancer Research Center, Heidelberg, Germany.,Institute of Reconstructive Neurobiology, School of Medicine & University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Anne Hoffrichter
- Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany.,Hector Institute for Translational Brain Research, Mannheim, Germany.,German Cancer Research Center, Heidelberg, Germany
| | - Ana Uzquiano
- INSERM U 1270, Paris, France.,Sorbonne Université, UMR-S 1270, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Fabio Marsoner
- Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany.,Hector Institute for Translational Brain Research, Mannheim, Germany.,German Cancer Research Center, Heidelberg, Germany
| | - Ruven Wilkens
- Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany.,Hector Institute for Translational Brain Research, Mannheim, Germany.,German Cancer Research Center, Heidelberg, Germany
| | - Marco Siekmann
- Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany.,Hector Institute for Translational Brain Research, Mannheim, Germany.,German Cancer Research Center, Heidelberg, Germany
| | - Bettina Bohl
- Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany.,Hector Institute for Translational Brain Research, Mannheim, Germany.,German Cancer Research Center, Heidelberg, Germany
| | - Andrea C Rossetti
- Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany.,Hector Institute for Translational Brain Research, Mannheim, Germany.,German Cancer Research Center, Heidelberg, Germany
| | - Sandra Horschitz
- Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany.,Hector Institute for Translational Brain Research, Mannheim, Germany.,German Cancer Research Center, Heidelberg, Germany
| | - Philipp Koch
- Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany.,Hector Institute for Translational Brain Research, Mannheim, Germany.,German Cancer Research Center, Heidelberg, Germany
| | - Fiona Francis
- INSERM U 1270, Paris, France.,Sorbonne Université, UMR-S 1270, Paris, France.,Institut du Fer à Moulin, Paris, France
| | - Julia Ladewig
- Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany.,Hector Institute for Translational Brain Research, Mannheim, Germany.,German Cancer Research Center, Heidelberg, Germany.,Institute of Reconstructive Neurobiology, School of Medicine & University Hospital Bonn, University of Bonn, Bonn, Germany
| |
Collapse
|
17
|
Shinmyo Y, Hamabe-Horiike T, Saito K, Kawasaki H. Investigation of the Mechanisms Underlying the Development and Evolution of the Cerebral Cortex Using Gyrencephalic Ferrets. Front Cell Dev Biol 2022; 10:847159. [PMID: 35386196 PMCID: PMC8977464 DOI: 10.3389/fcell.2022.847159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
The mammalian cerebral cortex has changed significantly during evolution. As a result of the increase in the number of neurons and glial cells in the cerebral cortex, its size has markedly expanded. Moreover, folds, called gyri and sulci, appeared on its surface, and its neuronal circuits have become much more complicated. Although these changes during evolution are considered to have been crucial for the acquisition of higher brain functions, the mechanisms underlying the development and evolution of the cerebral cortex of mammals are still unclear. This is, at least partially, because it is difficult to investigate these mechanisms using mice only. Therefore, genetic manipulation techniques for the cerebral cortex of gyrencephalic carnivore ferrets were developed recently. Furthermore, gene knockout was achieved in the ferret cerebral cortex using the CRISPR/Cas9 system. These techniques enabled molecular investigations using the ferret cerebral cortex. In this review, we will summarize recent findings regarding the mechanisms underlying the development and evolution of the mammalian cerebral cortex, mainly focusing on research using ferrets.
Collapse
Affiliation(s)
- Yohei Shinmyo
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Toshihide Hamabe-Horiike
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Kengo Saito
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
18
|
Ossola C, Kalebic N. Roots of the Malformations of Cortical Development in the Cell Biology of Neural Progenitor Cells. Front Neurosci 2022; 15:817218. [PMID: 35069108 PMCID: PMC8766818 DOI: 10.3389/fnins.2021.817218] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/14/2021] [Indexed: 12/13/2022] Open
Abstract
The cerebral cortex is a structure that underlies various brain functions, including cognition and language. Mammalian cerebral cortex starts developing during the embryonic period with the neural progenitor cells generating neurons. Newborn neurons migrate along progenitors’ radial processes from the site of their origin in the germinal zones to the cortical plate, where they mature and integrate in the forming circuitry. Cell biological features of neural progenitors, such as the location and timing of their mitoses, together with their characteristic morphologies, can directly or indirectly regulate the abundance and the identity of their neuronal progeny. Alterations in the complex and delicate process of cerebral cortex development can lead to malformations of cortical development (MCDs). They include various structural abnormalities that affect the size, thickness and/or folding pattern of the developing cortex. Their clinical manifestations can entail a neurodevelopmental disorder, such as epilepsy, developmental delay, intellectual disability, or autism spectrum disorder. The recent advancements of molecular and neuroimaging techniques, along with the development of appropriate in vitro and in vivo model systems, have enabled the assessment of the genetic and environmental causes of MCDs. Here we broadly review the cell biological characteristics of neural progenitor cells and focus on those features whose perturbations have been linked to MCDs.
Collapse
|
19
|
Terry BK, Park R, Cho SH, Crino PB, Kim S. Abnormal activation of Yap/Taz contributes to the pathogenesis of tuberous sclerosis complex. Hum Mol Genet 2022; 31:1979-1996. [PMID: 34999833 PMCID: PMC9239747 DOI: 10.1093/hmg/ddab374] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/31/2021] [Accepted: 12/27/2021] [Indexed: 01/09/2023] Open
Abstract
The multi-systemic genetic disorder tuberous sclerosis complex (TSC) impacts multiple neurodevelopmental processes including neuronal morphogenesis, neuronal migration, myelination and gliogenesis. These alterations contribute to the development of cerebral cortex abnormalities and malformations. Although TSC is caused by mTORC1 hyperactivation, cognitive and behavioral impairments are not improved through mTORC1 targeting, making the study of the downstream effectors of this complex important for understanding the mechanisms underlying TSC. As mTORC1 has been shown to promote the activity of the transcriptional co-activator Yap, we hypothesized that altered Yap/Taz signaling contributes to the pathogenesis of TSC. We first observed that the levels of Yap/Taz are increased in human cortical tuber samples and in embryonic cortices of Tsc2 conditional knockout (cKO) mice. Next, to determine how abnormal upregulation of Yap/Taz impacts the neuropathology of TSC, we deleted Yap/Taz in Tsc2 cKO mice. Importantly, Yap/Taz/Tsc2 triple conditional knockout (tcKO) animals show reduced cortical thickness and cortical neuron cell size, despite the persistence of high mTORC1 activity, suggesting that Yap/Taz play a downstream role in cytomegaly. Furthermore, Yap/Taz/Tsc2 tcKO significantly restored cortical and hippocampal lamination defects and reduced hippocampal heterotopia formation. Finally, the loss of Yap/Taz increased the distribution of myelin basic protein in Tsc2 cKO animals, consistent with an improvement in myelination. Overall, our results indicate that targeting Yap/Taz lessens the severity of neuropathology in a TSC animal model. This study is the first to implicate Yap/Taz as contributors to cortical pathogenesis in TSC and therefore as potential novel targets in the treatment of this disorder.
Collapse
Affiliation(s)
- Bethany K Terry
- Department of Neural Sciences, Lewis Katz School of Medicine, Shriners Hospitals Pediatrics Research Center, Temple University, Philadelphia, PA 19140, USA,Biomedical Sciences Graduate Program, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Raehee Park
- Department of Neural Sciences, Lewis Katz School of Medicine, Shriners Hospitals Pediatrics Research Center, Temple University, Philadelphia, PA 19140, USA
| | - Seo-Hee Cho
- Department of Medicine, Sidney Kimmel Medical College, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Peter B Crino
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Seonhee Kim
- To whom correspondence should be addressed. Tel: 215-926-9360; Fax: 215-926-9325;
| |
Collapse
|
20
|
Su YC, Hung TH, Wang TF, Lee YH, Wang TW, Yu JY. YAP maintains the production of intermediate progenitor cells and upper-layer projection neurons in the mouse cerebral cortex. Dev Dyn 2021; 251:846-863. [PMID: 34931379 DOI: 10.1002/dvdy.448] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 12/09/2021] [Accepted: 12/13/2021] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND The Hippo pathway is conserved through evolution and plays critical roles in development, tissue homeostasis and tumorigenesis. Yes-associated protein (YAP) is a transcriptional coactivator downstream of the Hippo pathway. Previous studies have demonstrated that activation of YAP promotes proliferation in the developing brain. Whether YAP is required for the production of neural progenitor cells or neurons in vivo remains unclear. RESULTS We demonstrated that SATB homeobox 2 (SATB2)-positive projection neurons (PNs) in upper layers, but not T-box brain transcription factor 1-positive and Coup-TF interacting protein 2-positive PNs in deep layers, were decreased in the neonatal cerebral cortex of Yap conditional knockout (cKO) mice driven by Nestin-Cre. Cell proliferation was reduced in the developing cerebral cortex of Yap-cKO. SATB2-positive PNs are largely generated from intermediate progenitor cells (IPCs), which are derived from radial glial cells (RGCs) during cortical development. Among these progenitor cells, IPCs but not RGCs were decreased in Yap-cKO. We further demonstrated that cell cycle re-entry was reduced in progenitor cells of Yap-cKO, suggesting that fewer IPCs were generated in Yap-cKO. CONCLUSION YAP is required for the production of IPCs and upper-layer SATB2-positive PNs during development of the cerebral cortex in mice.
Collapse
Affiliation(s)
- Yi-Ching Su
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tzu-Heng Hung
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tzu-Fang Wang
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ying-Hsuan Lee
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tsu-Wei Wang
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Jenn-Yah Yu
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
21
|
Gong P, Zou Y, Zhang W, Tian Q, Han S, Xu Z, Chen Q, Wang X, Li M. The neuroprotective effects of Insulin-Like Growth Factor 1 via the Hippo/YAP signaling pathway are mediated by the PI3K/AKT cascade following cerebral ischemia/reperfusion injury. Brain Res Bull 2021; 177:373-387. [PMID: 34717965 DOI: 10.1016/j.brainresbull.2021.10.017] [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: 02/07/2021] [Revised: 10/07/2021] [Accepted: 10/26/2021] [Indexed: 10/20/2022]
Abstract
Insulin-like growth factor 1 (IGF-1) has neuroprotective actions, including vasodilatory, anti-inflammatory, and antithrombotic effects, following ischemic stroke. However, the molecular mechanisms underlying the neuroprotective effects of IGF-1 following ischemic stroke remain unknown. Therefore, in the present study, we investigated whether IGF-1 exerted its neuroprotective effects by regulating the Hippo/YAP signaling pathway, potentially via activation of the PI3K/AKT cascade, following ischemic stroke. In the in vitro study, we exposed cultured PC12 and SH-5YSY cells, and cortical primary neurons, to oxygen-glucose deprivation. Cell viability was measured using CCK-8 assay. In the in vivo study, Sprague-Dawley rats were subjected to middle cerebral artery occlusion. Neurological function was assessed using a modified neurologic scoring system and the modified neurological severity score (mNSS) test, brain edema was detected by brain water content measurement, infarct volume was measured using triphenyltetrazolium chloride staining, and neuronal death and apoptosis were evaluated by TUNEL/NeuN double staining, HE and Nissl staining, and immunohistochemistry staining for NeuN. Finally, western blot analysis was used to measure the level of IGF-1 in vivo and levels of YAP/TAZ, PI3K and phosphorylated AKT (p-AKT) both in vitro and in vivo. IGF-1 induced activation of YAP/TAZ, which resulted in improved cell viability in vitro, and reduced neurological deficits, brain water content, neuronal death and apoptosis, and cerebral infarct volume in vivo. Notably, the neuroprotective effects of IGF-1 were blocked by an inhibitor of the PI3K/AKT cascade, LY294002. LY294002 treatment not only downregulated PI3K and p-AKT, but YAP/TAZ as well, leading to aggravation of neurological dysfunction and worsening of brain damage. Our findings indicate that the neuroprotective effects of IGF-1 are, at least in part mediated by upregulation of YAP/TAZ via activation of the PI3K/AKT cascade following cerebral ischemic stroke.
Collapse
Affiliation(s)
- Pian Gong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Yichun Zou
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, PR China
| | - Wei Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Qi Tian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Shoumeng Han
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Zhou Xu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Xin Wang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mingchang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China.
| |
Collapse
|
22
|
Engel-Pizcueta C, Pujades C. Interplay Between Notch and YAP/TAZ Pathways in the Regulation of Cell Fate During Embryo Development. Front Cell Dev Biol 2021; 9:711531. [PMID: 34490262 PMCID: PMC8417249 DOI: 10.3389/fcell.2021.711531] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/02/2021] [Indexed: 12/23/2022] Open
Abstract
Cells in growing tissues receive both biochemical and physical cues from their microenvironment. Growing evidence has shown that mechanical signals are fundamental regulators of cell behavior. However, how physical properties of the microenvironment are transduced into critical cell behaviors, such as proliferation, progenitor maintenance, or differentiation during development, is still poorly understood. The transcriptional co-activators YAP/TAZ shuttle between the cytoplasm and the nucleus in response to multiple inputs and have emerged as important regulators of tissue growth and regeneration. YAP/TAZ sense and transduce physical cues, such as those from the extracellular matrix or the actomyosin cytoskeleton, to regulate gene expression, thus allowing them to function as gatekeepers of progenitor behavior in several developmental contexts. The Notch pathway is a key signaling pathway that controls binary cell fate decisions through cell-cell communication in a context-dependent manner. Recent reports now suggest that the crosstalk between these two pathways is critical for maintaining the balance between progenitor maintenance and cell differentiation in different tissues. How this crosstalk integrates with morphogenesis and changes in tissue architecture during development is still an open question. Here, we discuss how progenitor cell proliferation, specification, and differentiation are coordinated with morphogenesis to construct a functional organ. We will pay special attention to the interplay between YAP/TAZ and Notch signaling pathways in determining cell fate decisions and discuss whether this represents a general mechanism of regulating cell fate during development. We will focus on research carried out in vertebrate embryos that demonstrate the important roles of mechanical cues in stem cell biology and discuss future challenges.
Collapse
Affiliation(s)
- Carolyn Engel-Pizcueta
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Cristina Pujades
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
23
|
Kalebic N, Namba T. Inheritance and flexibility of cell polarity: a clue for understanding human brain development and evolution. Development 2021; 148:272121. [PMID: 34499710 PMCID: PMC8451944 DOI: 10.1242/dev.199417] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cell polarity is fundamentally important for understanding brain development. Here, we hypothesize that the inheritance and flexibility of cell polarity during neocortex development could be implicated in neocortical evolutionary expansion. Molecular and morphological features of cell polarity may be inherited from one type of progenitor cell to the other and finally transmitted to neurons. Furthermore, key cell types, such as basal progenitors and neurons, exhibit a highly flexible polarity. We suggest that both inheritance and flexibility of cell polarity are implicated in the amplification of basal progenitors and tangential dispersion of neurons, which are key features of the evolutionary expansion of the neocortex. Summary: We suggest that the inheritance and flexibility of cell polarity are implicated in the evolutionary expansion of the developing neocortex by promoting the amplification of neural progenitors and tangential migration of neurons.
Collapse
Affiliation(s)
| | - Takashi Namba
- Neuroscience Center, HiLIFE - Helsinki Institute of Life Science, University of Helsinki, 00290 Helsinki, Finland
| |
Collapse
|
24
|
Lavado A, Gangwar R, Paré J, Wan S, Fan Y, Cao X. YAP/TAZ maintain the proliferative capacity and structural organization of radial glial cells during brain development. Dev Biol 2021; 480:39-49. [PMID: 34419458 DOI: 10.1016/j.ydbio.2021.08.010] [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: 05/05/2021] [Revised: 07/29/2021] [Accepted: 08/16/2021] [Indexed: 12/24/2022]
Abstract
The Hippo pathway regulates the development and homeostasis of many tissues and in many species. It controls the activity of two paralogous transcriptional coactivators, YAP and TAZ (YAP/TAZ). Although previous studies have established that aberrant YAP/TAZ activation is detrimental to mammalian brain development, whether and how endogenous levels of YAP/TAZ activity regulate brain development remain unclear. Here, we show that during mammalian cortical development, YAP/TAZ are specifically expressed in apical neural progenitor cells known as radial glial cells (RGCs). The subcellular localization of YAP/TAZ undergoes dynamic changes as corticogenesis proceeds. YAP/TAZ are required for maintaining the proliferative potential and structural organization of RGCs, and their ablation during cortical development reduces the numbers of cortical projection neurons and causes the loss of ependymal cells, resulting in hydrocephaly. Transcriptomic analysis using sorted RGCs reveals gene expression changes in YAP/TAZ-depleted cells that correlate with mutant phenotypes. Thus, our study has uncovered essential functions of YAP/TAZ during mammalian brain development and revealed the transcriptional mechanism of their action.
Collapse
Affiliation(s)
- Alfonso Lavado
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Ruchika Gangwar
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Joshua Paré
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Shibiao Wan
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Yiping Fan
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Xinwei Cao
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| |
Collapse
|
25
|
Pinson A, Huttner WB. Neocortex expansion in development and evolution-from genes to progenitor cell biology. Curr Opin Cell Biol 2021; 73:9-18. [PMID: 34098196 DOI: 10.1016/j.ceb.2021.04.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 04/30/2021] [Indexed: 12/12/2022]
Abstract
The evolutionary expansion of the neocortex, the seat of higher cognitive functions in humans, is primarily due to an increased and prolonged proliferation of neural progenitor cells during development. Basal progenitors, and in particular basal radial glial cells, are thought to have a key role in the increased generation of neurons that constitutes a foundation of neocortex expansion. Recent studies have identified primate-specific and human-specific genes and changes in gene expression that promote increased proliferative capacity of cortical progenitors. In many cases, the cell biological basis underlying this increase has been uncovered. Model systems such as mouse, ferret, nonhuman primates, and cerebral organoids have been used to establish the relevance of these genes for neocortex expansion.
Collapse
Affiliation(s)
- Anneline Pinson
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany.
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany.
| |
Collapse
|
26
|
Namba T, Haffner C, Huttner WB. Ex vivo Tissue Culture Protocols for Studying the Developing Neocortex. Bio Protoc 2021; 11:e4031. [PMID: 34150938 DOI: 10.21769/bioprotoc.4031] [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: 09/25/2020] [Revised: 02/19/2021] [Accepted: 02/25/2021] [Indexed: 11/02/2022] Open
Abstract
The size of the neocortex and its morphology are highly divergent across mammalian species. Several approaches have been utilized for the analysis of neocortical development and comparison among different species. In the present protocol (Note: This protocol requires basic knowledge of brain anatomy), we describe three ex vivo neocortical slice/tissue culture methods: (i) organotypic slice culture (mouse, ferret, human); (ii) hemisphere rotation culture (mouse, ferret); and (iii) free-floating tissue culture (mouse, ferret, human). Each of these three culture methods offers distinct features with regard to the analyses to be performed and can be combined with genetic manipulation by electroporation and treatment with specific inhibitors. These three culture methods are therefore powerful techniques to examine the function of genes involved in neocortical development.
Collapse
Affiliation(s)
- Takashi Namba
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany.,Neuroscience Center, HiLIFE - Helsinki Institute of Life Science, University of Helsinki, FI-00014 Helsinki, Finland
| | - Christiane Haffner
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| |
Collapse
|
27
|
Xing L, Kubik-Zahorodna A, Namba T, Pinson A, Florio M, Prochazka J, Sarov M, Sedlacek R, Huttner WB. Expression of human-specific ARHGAP11B in mice leads to neocortex expansion and increased memory flexibility. EMBO J 2021; 40:e107093. [PMID: 33938018 PMCID: PMC8246068 DOI: 10.15252/embj.2020107093] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 03/20/2021] [Accepted: 03/25/2021] [Indexed: 12/14/2022] Open
Abstract
Neocortex expansion during human evolution provides a basis for our enhanced cognitive abilities. Yet, which genes implicated in neocortex expansion are actually responsible for higher cognitive abilities is unknown. The expression of human-specific ARHGAP11B in embryonic/foetal mouse, ferret and marmoset neocortex was previously found to promote basal progenitor proliferation, upper-layer neuron generation and neocortex expansion during development, features commonly thought to contribute to increased cognitive abilities. However, a key question is whether this phenotype persists into adulthood and if so, whether cognitive abilities are indeed increased. Here, we generated a transgenic mouse line with physiological ARHGAP11B expression that exhibits increased neocortical size and upper-layer neuron numbers persisting into adulthood. Adult ARHGAP11B-transgenic mice showed altered neurobehaviour, notably increased memory flexibility and a reduced anxiety level. Our data are consistent with the notion that neocortex expansion by ARHGAP11B, a gene implicated in human evolution, underlies some of the altered neurobehavioural features observed in the transgenic mice, such as the increased memory flexibility, a neocortex-associated trait, with implications for the increase in cognitive abilities during human evolution.
Collapse
Affiliation(s)
- Lei Xing
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Agnieszka Kubik-Zahorodna
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Takashi Namba
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Anneline Pinson
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Marta Florio
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Jan Prochazka
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Mihail Sarov
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Radislav Sedlacek
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
28
|
Gilardi C, Kalebic N. The Ferret as a Model System for Neocortex Development and Evolution. Front Cell Dev Biol 2021; 9:661759. [PMID: 33996819 PMCID: PMC8118648 DOI: 10.3389/fcell.2021.661759] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/01/2021] [Indexed: 12/19/2022] Open
Abstract
The neocortex is the largest part of the cerebral cortex and a key structure involved in human behavior and cognition. Comparison of neocortex development across mammals reveals that the proliferative capacity of neural stem and progenitor cells and the length of the neurogenic period are essential for regulating neocortex size and complexity, which in turn are thought to be instrumental for the increased cognitive abilities in humans. The domesticated ferret, Mustela putorius furo, is an important animal model in neurodevelopment for its complex postnatal cortical folding, its long period of forebrain development and its accessibility to genetic manipulation in vivo. Here, we discuss the molecular, cellular, and histological features that make this small gyrencephalic carnivore a suitable animal model to study the physiological and pathological mechanisms for the development of an expanded neocortex. We particularly focus on the mechanisms of neural stem cell proliferation, neuronal differentiation, cortical folding, visual system development, and neurodevelopmental pathologies. We further discuss the technological advances that have enabled the genetic manipulation of the ferret in vivo. Finally, we compare the features of neocortex development in the ferret with those of other model organisms.
Collapse
|
29
|
Ulmke PA, Sakib MS, Ditte P, Sokpor G, Kerimoglu C, Pham L, Xie Y, Mao X, Rosenbusch J, Teichmann U, Nguyen HP, Fischer A, Eichele G, Staiger JF, Tuoc T. Molecular Profiling Reveals Involvement of ESCO2 in Intermediate Progenitor Cell Maintenance in the Developing Mouse Cortex. Stem Cell Reports 2021; 16:968-984. [PMID: 33798452 PMCID: PMC8072132 DOI: 10.1016/j.stemcr.2021.03.008] [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: 08/13/2020] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Intermediate progenitor cells (IPCs) are neocortical neuronal precursors. Although IPCs play crucial roles in corticogenesis, their molecular features remain largely unknown. In this study, we aimed to characterize the molecular profile of IPCs. We isolated TBR2-positive (+) IPCs and TBR2-negative (-) cell populations in the developing mouse cortex. Comparative genome-wide gene expression analysis of TBR2+ IPCs versus TBR2- cells revealed differences in key factors involved in chromatid segregation, cell-cycle regulation, transcriptional regulation, and cell signaling. Notably, mutation of many IPC genes in human has led to intellectual disability and caused a wide range of cortical malformations, including microcephaly and agenesis of corpus callosum. Loss-of-function experiments in cortex-specific mutants of Esco2, one of the novel IPC genes, demonstrate its critical role in IPC maintenance, and substantiate the identification of a central genetic determinant of IPC biogenesis. Our data provide novel molecular characteristics of IPCs in the developing mouse cortex.
Collapse
Affiliation(s)
- Pauline Antonie Ulmke
- Institute for Neuroanatomy, University Medical Center, Georg-August-University, Goettingen, Germany
| | - M Sadman Sakib
- German Center for Neurodegenerative Diseases, Goettingen, Germany
| | - Peter Ditte
- Max-Planck-Institute for Biophysical Chemistry, Goettingen, Germany
| | - Godwin Sokpor
- Institute for Neuroanatomy, University Medical Center, Georg-August-University, Goettingen, Germany; Department of Human Genetics, Ruhr University of Bochum, Bochum, Germany
| | - Cemil Kerimoglu
- German Center for Neurodegenerative Diseases, Goettingen, Germany
| | - Linh Pham
- Institute for Neuroanatomy, University Medical Center, Georg-August-University, Goettingen, Germany; Department of Human Genetics, Ruhr University of Bochum, Bochum, Germany
| | - Yuanbin Xie
- Institute for Neuroanatomy, University Medical Center, Georg-August-University, Goettingen, Germany
| | - Xiaoyi Mao
- Institute for Neuroanatomy, University Medical Center, Georg-August-University, Goettingen, Germany
| | - Joachim Rosenbusch
- Institute for Neuroanatomy, University Medical Center, Georg-August-University, Goettingen, Germany
| | - Ulrike Teichmann
- Max-Planck-Institute for Biophysical Chemistry, Goettingen, Germany
| | - Huu Phuc Nguyen
- Department of Human Genetics, Ruhr University of Bochum, Bochum, Germany
| | - Andre Fischer
- German Center for Neurodegenerative Diseases, Goettingen, Germany; Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), Goettingen, Germany
| | - Gregor Eichele
- Max-Planck-Institute for Biophysical Chemistry, Goettingen, Germany
| | - Jochen F Staiger
- Institute for Neuroanatomy, University Medical Center, Georg-August-University, Goettingen, Germany
| | - Tran Tuoc
- Institute for Neuroanatomy, University Medical Center, Georg-August-University, Goettingen, Germany; Department of Human Genetics, Ruhr University of Bochum, Bochum, Germany.
| |
Collapse
|
30
|
Hu HB, Song ZQ, Song GP, Li S, Tu HQ, Wu M, Zhang YC, Yuan JF, Li TT, Li PY, Xu YL, Shen XL, Han QY, Li AL, Zhou T, Chun J, Zhang XM, Li HY. LPA signaling acts as a cell-extrinsic mechanism to initiate cilia disassembly and promote neurogenesis. Nat Commun 2021; 12:662. [PMID: 33510165 PMCID: PMC7843646 DOI: 10.1038/s41467-021-20986-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 12/15/2020] [Indexed: 01/17/2023] Open
Abstract
Dynamic assembly and disassembly of primary cilia controls embryonic development and tissue homeostasis. Dysregulation of ciliogenesis causes human developmental diseases termed ciliopathies. Cell-intrinsic regulatory mechanisms of cilia disassembly have been well-studied. The extracellular cues controlling cilia disassembly remain elusive, however. Here, we show that lysophosphatidic acid (LPA), a multifunctional bioactive phospholipid, acts as a physiological extracellular factor to initiate cilia disassembly and promote neurogenesis. Through systematic analysis of serum components, we identify a small molecular-LPA as the major driver of cilia disassembly. Genetic inactivation and pharmacological inhibition of LPA receptor 1 (LPAR1) abrogate cilia disassembly triggered by serum. The LPA-LPAR-G-protein pathway promotes the transcription and phosphorylation of cilia disassembly factors-Aurora A, through activating the transcription coactivators YAP/TAZ and calcium/CaM pathway, respectively. Deletion of Lpar1 in mice causes abnormally elongated cilia and decreased proliferation in neural progenitor cells, thereby resulting in defective neurogenesis. Collectively, our findings establish LPA as a physiological initiator of cilia disassembly and suggest targeting the metabolism of LPA and the LPA pathway as potential therapies for diseases with dysfunctional ciliogenesis.
Collapse
Affiliation(s)
- Huai-Bin Hu
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Zeng-Qing Song
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Guang-Ping Song
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Sen Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Hai-Qing Tu
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Min Wu
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Yu-Cheng Zhang
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Jin-Feng Yuan
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Ting-Ting Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Pei-Yao Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Yu-Ling Xu
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Xiao-Lin Shen
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Qiu-Ying Han
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Ai-Ling Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Tao Zhou
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Jerold Chun
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, USA
| | - Xue-Min Zhang
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China.
| | - Hui-Yan Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China.
- School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
31
|
Abstract
The mammalian cerebral cortex is the pinnacle of brain evolution, reaching its maximum complexity in terms of neuron number, diversity and functional circuitry. The emergence of this outstanding complexity begins during embryonic development, when a limited number of neural stem and progenitor cells manage to generate myriads of neurons in the appropriate numbers, types and proportions, in a process called neurogenesis. Here we review the current knowledge on the regulation of cortical neurogenesis, beginning with a description of the types of progenitor cells and their lineage relationships. This is followed by a review of the determinants of neuron fate, the molecular and genetic regulatory mechanisms, and considerations on the evolution of cortical neurogenesis in vertebrates leading to humans. We finish with an overview on how dysregulation of neurogenesis is a leading cause of human brain malformations and functional disabilities.
Collapse
Affiliation(s)
- Ana Villalba
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant, Spain
| | - Magdalena Götz
- Institute for Stem Cell Research, Helmholtz Zentrum München & Biomedical Center, Ludwig-Maximilians Universitaet, Planegg-Martinsried, Germany
| | - Víctor Borrell
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant, Spain.
| |
Collapse
|
32
|
Nishizono H, Yasuda R, Laviv T. Methodologies and Challenges for CRISPR/Cas9 Mediated Genome Editing of the Mammalian Brain. Front Genome Ed 2020; 2:602970. [PMID: 34713226 PMCID: PMC8525404 DOI: 10.3389/fgeed.2020.602970] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/06/2020] [Indexed: 01/22/2023] Open
Abstract
Neurons and glia are highly polarized cells with extensive subcellular structures extending over large distances from their cell bodies. Previous research has revealed elaborate protein signaling complexes localized within intracellular compartments. Thus, exploring the function and the localization of endogenous proteins is vital to understanding the precise molecular mechanisms underlying the synapse, cellular, and circuit function. Recent advances in CRISPR/Cas9-based genome editing techniques have allowed researchers to rapidly develop transgenic animal models and perform single-cell level genome editing in the mammalian brain. Here, we introduce and comprehensively review the latest techniques for genome-editing in whole animals using fertilized eggs and methods for gene editing in specific neuronal populations in the adult or developing mammalian brain. Finally, we describe the advantages and disadvantages of each technique, as well as the challenges that lie ahead to advance the generation of methodologies for genome editing in the brain using the current CRISPR/Cas9 system.
Collapse
Affiliation(s)
- Hirofumi Nishizono
- Department of Neuronal Signal Transduction, Max Planck Florida Institute for Neuroscience, Jupiter, FL, United States
| | - Ryohei Yasuda
- Department of Neuronal Signal Transduction, Max Planck Florida Institute for Neuroscience, Jupiter, FL, United States
| | | |
Collapse
|
33
|
Xing L, Kalebic N, Namba T, Vaid S, Wimberger P, Huttner WB. Serotonin Receptor 2A Activation Promotes Evolutionarily Relevant Basal Progenitor Proliferation in the Developing Neocortex. Neuron 2020; 108:1113-1129.e6. [PMID: 33080227 DOI: 10.1016/j.neuron.2020.09.034] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 07/17/2020] [Accepted: 09/24/2020] [Indexed: 12/26/2022]
Abstract
Evolutionary expansion of the mammalian neocortex (Ncx) has been linked to increased abundance and proliferative capacity of basal progenitors (BPs) in the subventricular zone during development. BP proliferation is governed by both intrinsic and extrinsic signals, several of which have been identified. However, a role of neurotransmitters, a canonical class of extrinsic signaling molecules, in BP proliferation remains to be established. Here, we show that serotonin (5-HT), via its receptor HTR2A, promotes BP proliferation in an evolutionarily relevant manner. HTR2A is not expressed in embryonic mouse Ncx; accordingly, 5-HT does not increase mouse BP proliferation. However, ectopic HTR2A expression can increase mouse BP proliferation. Conversely, CRISPR/Cas9-mediated knockout of endogenous HTR2A in embryonic ferret Ncx reduces BP proliferation. Pharmacological activation of endogenous HTR2A in fetal human Ncx ex vivo increases BP proliferation via HER2/ERK signaling. Hence, 5-HT emerges as an important extrinsic pro-proliferative signal for BPs, which may have contributed to evolutionary Ncx expansion.
Collapse
Affiliation(s)
- Lei Xing
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
| | - Nereo Kalebic
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany; Human Technopole, Via Cristina Belgioioso 171, Milan, Italy
| | - Takashi Namba
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
| | - Samir Vaid
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
| | - Pauline Wimberger
- Technische Universität Dresden, Universitätsklinikum Carl Gustav Carus, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Dresden, Germany
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany.
| |
Collapse
|
34
|
Modeling human neuronal migration deficits in 3D. Curr Opin Neurobiol 2020; 66:30-36. [PMID: 33069990 DOI: 10.1016/j.conb.2020.09.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/23/2020] [Accepted: 09/02/2020] [Indexed: 10/23/2022]
Abstract
During the past few decades, we have witnessed an impressive gain in the knowledge regarding the basic mechanisms underlying human neuronal migration disorders by the usage of mouse models. Nevertheless, despite the remarkable conservation both in the genetic encoded information and the developmental processes, there are still numerous important differences between human and mouse. This may explain the vast excitement following the realization that technological breakthroughs enabled generating tissue-like human-based organoids for modeling human neuronal migration diseases. This review will provide a short introduction on human and mouse neuronal migration processes, and highlight human brain organoid models of neuronal migration diseases.
Collapse
|
35
|
Han D, Lee SM, Kwon M, Noh H, Lee JH, Yoon Y, Cho JY, Yoon K. YAP Enhances FGF2-Dependent Neural Stem Cell Proliferation by Induction of FGF Receptor Expression. Stem Cells Dev 2020; 29:1240-1246. [PMID: 32669047 DOI: 10.1089/scd.2019.0281] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The Hippo signaling pathway regulates cell proliferation and organ growth, and its activation is mainly reflected by the phosphorylation levels of Yes-associated protein (YAP). In this study, we show that YAP facilitates embryonic neural stem cell proliferation by elevating their responsiveness to fibroblast growth factor 2 (FGF2), one of the major growth factors for neural stem cells, in vivo as well as in vitro. Western blot and quantitative real-time PCR analyses revealed that expression of the FGF receptors (FGFRs) FGFR1 to FGFR4 were greatly increased by YAP expression upon FGF2 treatment, followed by upregulation of the mitogen-activated protein kinase and protein kinase B signaling pathways. Furthermore, as assessed by quantitative real-time PCR analyses, YAP-induced FGFR expression was found to be TEA domain transcription factor (TEAD)-independent, and transcriptional coactivator with PDZ-binding motif, the other homolog of Yorki in the Drosophila Hippo signaling pathway, was found to possess similar activity to YAP. Finally, adjustment of FGFR signaling activity in the YAP-expressing cells to control levels efficiently offset the cell proliferative effects of YAP, suggesting that the increased proliferation of YAP-expressing neural stem cells was mainly attributable to enhanced FGFR signaling. Our data indicate that YAP plays an important role in neural stem cell regulation by elevating FGFR expression, subsequently leading to enhanced cell proliferation.
Collapse
Affiliation(s)
- Dasol Han
- College of Biotechnology and Bioengineering, Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sun Min Lee
- College of Biotechnology and Bioengineering, Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Mookwang Kwon
- College of Biotechnology and Bioengineering, Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hogyun Noh
- College of Biotechnology and Bioengineering, Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ju Hyun Lee
- College of Biotechnology and Bioengineering, Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Youngik Yoon
- College of Biotechnology and Bioengineering, Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jae Youl Cho
- College of Biotechnology and Bioengineering, Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Keejung Yoon
- College of Biotechnology and Bioengineering, Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
36
|
Najas S, Pijuan I, Esteve-Codina A, Usieto S, Martinez JD, Zwijsen A, Arbonés ML, Martí E, Le Dréau G. A SMAD1/5-YAP signalling module drives radial glia self-amplification and growth of the developing cerebral cortex. Development 2020; 147:dev.187005. [PMID: 32541003 DOI: 10.1242/dev.187005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 06/02/2020] [Indexed: 12/19/2022]
Abstract
The growth and evolutionary expansion of the cerebral cortex are defined by the spatial-temporal production of neurons, which itself depends on the decision of radial glial cells (RGCs) to self-amplify or to switch to neurogenic divisions. The mechanisms regulating these RGC fate decisions are still incompletely understood. Here, we describe a novel and evolutionarily conserved role of the canonical BMP transcription factors SMAD1/5 in controlling neurogenesis and growth during corticogenesis. Reducing the expression of both SMAD1 and SMAD5 in neural progenitors at early mouse cortical development caused microcephaly and an increased production of early-born cortical neurons at the expense of late-born ones, which correlated with the premature differentiation and depletion of the pool of cortical progenitors. Gain- and loss-of-function experiments performed during early cortical neurogenesis in the chick revealed that SMAD1/5 activity supports self-amplifying RGC divisions and restrains the neurogenic ones. Furthermore, we demonstrate that SMAD1/5 stimulate RGC self-amplification through the positive post-transcriptional regulation of the Hippo signalling effector YAP. We anticipate this SMAD1/5-YAP signalling module to be fundamental in controlling growth and evolution of the amniote cerebral cortex.
Collapse
Affiliation(s)
- Sonia Najas
- Department of Developmental Biology, Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/ Baldiri Reixac 10-15, 08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - Isabel Pijuan
- Department of Developmental Biology, Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/ Baldiri Reixac 10-15, 08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - Anna Esteve-Codina
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 4, 08028 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Susana Usieto
- Department of Developmental Biology, Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/ Baldiri Reixac 10-15, 08028 Barcelona, Spain
| | - Juan D Martinez
- Department of Developmental Biology, Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/ Baldiri Reixac 10-15, 08028 Barcelona, Spain
| | - An Zwijsen
- Department of Cardiovascular Sciences, KU Leuven, Leuven 3000, Belgium
| | - Maria L Arbonés
- Department of Developmental Biology, Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/ Baldiri Reixac 10-15, 08028 Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - Elisa Martí
- Department of Developmental Biology, Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/ Baldiri Reixac 10-15, 08028 Barcelona, Spain
| | - Gwenvael Le Dréau
- Department of Developmental Biology, Instituto de Biología Molecular de Barcelona, CSIC, Parc Científic de Barcelona, C/ Baldiri Reixac 10-15, 08028 Barcelona, Spain
| |
Collapse
|
37
|
Vaid S, Huttner WB. Transcriptional Regulators and Human-Specific/Primate-Specific Genes in Neocortical Neurogenesis. Int J Mol Sci 2020; 21:ijms21134614. [PMID: 32610533 PMCID: PMC7369782 DOI: 10.3390/ijms21134614] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/09/2020] [Accepted: 06/26/2020] [Indexed: 12/15/2022] Open
Abstract
During development, starting from a pool of pluripotent stem cells, tissue-specific genetic programs help to shape and develop functional organs. To understand the development of an organ and its disorders, it is important to understand the spatio-temporal dynamics of the gene expression profiles that occur during its development. Modifications in existing genes, the de-novo appearance of new genes, or, occasionally, even the loss of genes, can greatly affect the gene expression profile of any given tissue and contribute to the evolution of organs or of parts of organs. The neocortex is evolutionarily the most recent part of the brain, it is unique to mammals, and is the seat of our higher cognitive abilities. Progenitors that give rise to this tissue undergo sequential waves of differentiation to produce the complete sets of neurons and glial cells that make up a functional neocortex. We will review herein our understanding of the transcriptional regulators that control the neural precursor cells (NPCs) during the generation of the most abundant class of neocortical neurons, the glutametergic neurons. In addition, we will discuss the roles of recently-identified human- and primate-specific genes in promoting neurogenesis, leading to neocortical expansion.
Collapse
|
38
|
Eder N, Roncaroli F, Domart MC, Horswell S, Andreiuolo F, Flynn HR, Lopes AT, Claxton S, Kilday JP, Collinson L, Mao JH, Pietsch T, Thompson B, Snijders AP, Ultanir SK. YAP1/TAZ drives ependymoma-like tumour formation in mice. Nat Commun 2020; 11:2380. [PMID: 32404936 PMCID: PMC7220953 DOI: 10.1038/s41467-020-16167-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 04/17/2020] [Indexed: 11/09/2022] Open
Abstract
YAP1 gene fusions have been observed in a subset of paediatric ependymomas. Here we show that, ectopic expression of active nuclear YAP1 (nlsYAP5SA) in ventricular zone neural progenitor cells using conditionally-induced NEX/NeuroD6-Cre is sufficient to drive brain tumour formation in mice. Neuronal differentiation is inhibited in the hippocampus. Deletion of YAP1's negative regulators LATS1 and LATS2 kinases in NEX-Cre lineage in double conditional knockout mice also generates similar tumours, which are rescued by deletion of YAP1 and its paralog TAZ. YAP1/TAZ-induced mouse tumours display molecular and ultrastructural characteristics of human ependymoma. RNA sequencing and quantitative proteomics of mouse tumours demonstrate similarities to YAP1-fusion induced supratentorial ependymoma. Finally, we find that transcriptional cofactor HOPX is upregulated in mouse models and in human YAP1-fusion induced ependymoma, supporting their similarity. Our results show that uncontrolled YAP1/TAZ activity in neuronal precursor cells leads to ependymoma-like tumours in mice.
Collapse
Affiliation(s)
- Noreen Eder
- Kinases and Brain Development Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
- Protein Analysis and Proteomics Platform, The Francis Crick Institute, London, NW1 1AT, UK
| | - Federico Roncaroli
- Manchester Centre for Clinical Neuroscience, Salford Royal NHS Foundation Trust, Salford and Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, School of Biology, University of Manchester, Manchester, M13 9PT, UK
| | | | - Stuart Horswell
- Bioinformatics and Biostatistics Platform, The Francis Crick Institute, London, NW1 1AT, UK
| | - Felipe Andreiuolo
- Institute of Neuropathology, DGNN Brain Tumour Reference Center, University of Bonn, Bonn, Germany
| | - Helen R Flynn
- Protein Analysis and Proteomics Platform, The Francis Crick Institute, London, NW1 1AT, UK
| | - Andre T Lopes
- Kinases and Brain Development Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Suzanne Claxton
- Kinases and Brain Development Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - John-Paul Kilday
- Centre for Paediatric, Teenage and Young Adult Cancer, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Lucy Collinson
- Electron Microscopy Platform, The Francis Crick Institute, London, NW1 1AT, UK
| | - Jun-Hao Mao
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Torsten Pietsch
- Institute of Neuropathology, DGNN Brain Tumour Reference Center, University of Bonn, Bonn, Germany
| | - Barry Thompson
- Epithelial Biology Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Ambrosius P Snijders
- Protein Analysis and Proteomics Platform, The Francis Crick Institute, London, NW1 1AT, UK
| | - Sila K Ultanir
- Kinases and Brain Development Laboratory, The Francis Crick Institute, London, NW1 1AT, UK.
| |
Collapse
|
39
|
Tead transcription factors differentially regulate cortical development. Sci Rep 2020; 10:4625. [PMID: 32170161 PMCID: PMC7070074 DOI: 10.1038/s41598-020-61490-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 02/27/2020] [Indexed: 12/16/2022] Open
Abstract
Neural stem cells (NSCs) generate neurons of the cerebral cortex with distinct morphologies and functions. How specific neuron production, differentiation and migration are orchestrated is unclear. Hippo signaling regulates gene expression through Tead transcription factors (TFs). We show that Hippo transcriptional coactivators Yap1/Taz and the Teads have distinct functions during cortical development. Yap1/Taz promote NSC maintenance and Satb2+ neuron production at the expense of Tbr1+ neuron generation. However, Teads have moderate effects on NSC maintenance and do not affect Satb2+ neuron differentiation. Conversely, whereas Tead2 blocks Tbr1+ neuron formation, Tead1 and Tead3 promote this early fate. In addition, we found that Hippo effectors regulate neuronal migration to the cortical plate (CP) in a reciprocal fashion, that ApoE, Dab2 and Cyr61 are Tead targets, and these contribute to neuronal fate determination and migration. Our results indicate that multifaceted Hippo signaling is pivotal in different aspects of cortical development.
Collapse
|
40
|
Thompson BJ. YAP/TAZ: Drivers of Tumor Growth, Metastasis, and Resistance to Therapy. Bioessays 2020; 42:e1900162. [DOI: 10.1002/bies.201900162] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 02/11/2020] [Indexed: 01/17/2023]
Affiliation(s)
- Barry J. Thompson
- EMBL AustraliaJohn Curtin School of Medical ResearchThe Australian National University 131 Garran Rd, Acton 2602 Canberra ACT Australia
| |
Collapse
|
41
|
Abstract
The neocortex is the largest part of the mammalian brain and is the seat of our higher cognitive functions. This outstanding neural structure increased massively in size and complexity during evolution in a process recapitulated today during the development of extant mammals. Accordingly, defects in neocortical development commonly result in severe intellectual and social deficits. Thus, understanding the development of the neocortex benefits from understanding its evolution and disease and also informs about their underlying mechanisms. Here, I briefly summarize the most recent and outstanding advances in our understanding of neocortical development and focus particularly on dorsal progenitors and excitatory neurons. I place special emphasis on the specification of neural stem cells in distinct classes and their proliferation and production of neurons and then discuss recent findings on neuronal migration. Recent discoveries on the genetic evolution of neocortical development are presented with a particular focus on primates. Progress on all these fronts is being accelerated by high-throughput gene expression analyses and particularly single-cell transcriptomics. I end with novel insights into the involvement of microglia in embryonic brain development and how improvements in cultured cerebral organoids are gradually consolidating them as faithful models of neocortex development in humans.
Collapse
Affiliation(s)
- Victor Borrell
- Institute of Neuroscience, Consejo Superior de Investigaciones Científicas (CSIC) - Universidad Miguel Hernández, Ramon y Cajal s/n, 03550 San Juan de Alicante, Spain
| |
Collapse
|
42
|
Sapir T, Barakat TS, Paredes MF, Lerman-Sagie T, Aronica E, Klonowski W, Nguyen L, Ben Zeev B, Bahi-Buisson N, Leventer R, Rachmian N, Reiner O. Building Bridges Between the Clinic and the Laboratory: A Meeting Review - Brain Malformations: A Roadmap for Future Research. Front Cell Neurosci 2019; 13:434. [PMID: 31611776 PMCID: PMC6776596 DOI: 10.3389/fncel.2019.00434] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 09/09/2019] [Indexed: 01/08/2023] Open
Abstract
In the middle of March 2019, a group of scientists and clinicians (as well as those who wear both hats) gathered in the green campus of the Weizmann Institute of Science to share recent scientific findings, to establish collaborations, and to discuss future directions for better diagnosis, etiology modeling and treatment of brain malformations. One hundred fifty scientists from twenty-two countries took part in this meeting. Thirty-eight talks were presented and as many as twenty-five posters were displayed. This review is aimed at presenting some of the highlights that the audience was exposed to during the three-day meeting.
Collapse
Affiliation(s)
- Tamar Sapir
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Tahsin Stefan Barakat
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Mercedes F. Paredes
- Department of Neurology and Neuroscience Graduate Division, University of California, San Francisco, San Francisco, CA, United States
| | - Tally Lerman-Sagie
- Pediatric Neurology Unit, Fetal Neurology Clinic, Wolfson Medical Center, Holon and Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eleonora Aronica
- Department of (Neuro-)Pathology, Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN), Zwolle, Netherlands
| | - Wlodzimierz Klonowski
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Laurent Nguyen
- GIGA-Stem Cells, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), C.H.U. Sart Tilman, University of Liège, Liège, Belgium
| | - Bruria Ben Zeev
- Sackler School of Medicine and Pediatric Neurology Unit, Edmond and Lilly Safra Pediatric Hospital, Tel Aviv University, Tel Aviv, Israel
| | - Nadia Bahi-Buisson
- INSERM UMR 1163, Imagine Institute, Paris Descartes University, Paris, France
- Necker Enfants Malades Hospital, Pediatrric Neurology APHP, Paris, France
| | - Richard Leventer
- Department of Neurology, Royal Children’s Hospital, Murdoch Children’s Research Institute, University of Melbourne, Parkville, VIC, Australia
- Department of Pediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Noa Rachmian
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Orly Reiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
43
|
Suzuki IK. Molecular drivers of human cerebral cortical evolution. Neurosci Res 2019; 151:1-14. [PMID: 31175883 DOI: 10.1016/j.neures.2019.05.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/30/2019] [Accepted: 05/31/2019] [Indexed: 01/10/2023]
Abstract
One of the most important questions in human evolutionary biology is how our ancestor has acquired an expanded volume of the cerebral cortex, which may have significantly impacted on improving our cognitive abilities. Recent comparative approaches have identified developmental features unique to the human or hominid cerebral cortex, not shared with other animals including conventional experimental models. In addition, genomic, transcriptomic, and epigenomic signatures associated with human- or hominid-specific processes of the cortical development are becoming identified by virtue of technical progress in the deep nucleotide sequencing. This review discusses ontogenic and phylogenetic processes of the human cerebral cortex, followed by the introduction of recent comprehensive approaches identifying molecular mechanisms potentially driving the evolutionary changes in the cortical development.
Collapse
Affiliation(s)
- Ikuo K Suzuki
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033 Japan; VIB-KU Leuven Center for Brain & Disease Research, 3000 Leuven, Belgium; Department of Neurosciences, Leuven Brain Institute, KULeuven, 3000 Leuven, Belgium; Université Libre de Bruxelles (U.L.B.), Institut de Recherches en Biologie Humaine et Moléculaire (IRIBHM), ULB Neuroscience Institute (UNI), 1070 Brussels, Belgium.
| |
Collapse
|
44
|
Roy A, Murphy RM, Deng M, MacDonald JW, Bammler TK, Aldinger KA, Glass IA, Millen KJ. PI3K-Yap activity drives cortical gyrification and hydrocephalus in mice. eLife 2019; 8:45961. [PMID: 31094678 PMCID: PMC6544437 DOI: 10.7554/elife.45961] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 05/15/2019] [Indexed: 01/07/2023] Open
Abstract
Mechanisms driving the initiation of brain folding are incompletely understood. We have previously characterized mouse models recapitulating human PIK3CA-related brain overgrowth, epilepsy, dysplastic gyrification and hydrocephalus (Roy et al., 2015). Using the same, highly regulatable brain-specific model, here we report PI3K-dependent mechanisms underlying gyrification of the normally smooth mouse cortex, and hydrocephalus. We demonstrate that a brief embryonic Pik3ca activation was sufficient to drive subtle changes in apical cell adhesion and subcellular Yap translocation, causing focal proliferation and subsequent initiation of the stereotypic ‘gyrification sequence’, seen in naturally gyrencephalic mammals. Treatment with verteporfin, a nuclear Yap inhibitor, restored apical surface integrity, normalized proliferation, attenuated gyrification and rescued the associated hydrocephalus, highlighting the interrelated role of regulated PI3K-Yap signaling in normal neural-ependymal development. Our data defines apical cell-adhesion as the earliest known substrate for cortical gyrification. In addition, our preclinical results support the testing of Yap-related small-molecule therapeutics for developmental hydrocephalus.
Collapse
Affiliation(s)
- Achira Roy
- Center for Integrative Brain Research, Seattle Children's Research Institute, Washington, United States
| | - Rory M Murphy
- Center for Integrative Brain Research, Seattle Children's Research Institute, Washington, United States
| | - Mei Deng
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Washington, United States
| | - James W MacDonald
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Washington, United States
| | - Theo K Bammler
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Washington, United States
| | - Kimberly A Aldinger
- Center for Integrative Brain Research, Seattle Children's Research Institute, Washington, United States.,Division of Genetic Medicine, Department of Pediatrics, University of Washington, Washington, United States
| | - Ian A Glass
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Washington, United States
| | - Kathleen J Millen
- Center for Integrative Brain Research, Seattle Children's Research Institute, Washington, United States.,Division of Genetic Medicine, Department of Pediatrics, University of Washington, Washington, United States
| |
Collapse
|