1
|
Vasan L, Chinchalongporn V, Saleh F, Zinyk D, Ke C, Suresh H, Ghazale H, Belfiore L, Touahri Y, Oproescu AM, Patel S, Rozak M, Amemiya Y, Han S, Moffat A, Black SE, McLaurin J, Near J, Seth A, Goubran M, Reiner O, Gillis J, Wang C, Okawa S, Schuurmans C. Examining the NEUROG2 lineage and associated gene expression in human cortical organoids. Development 2025; 152:dev202703. [PMID: 39680368 PMCID: PMC11829764 DOI: 10.1242/dev.202703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 12/07/2024] [Indexed: 12/17/2024]
Abstract
Proneural genes are conserved drivers of neurogenesis across the animal kingdom. How their functions have adapted to guide human-specific neurodevelopmental features is poorly understood. Here, we mined transcriptomic data from human fetal cortices and generated from human embryonic stem cell-derived cortical organoids (COs) to show that NEUROG1 and NEUROG2 are most highly expressed in basal neural progenitor cells, with pseudotime trajectory analyses indicating that NEUROG1-derived lineages predominate early and NEUROG2 lineages later. Using ChIP-qPCR, gene silencing and overexpression studies in COs, we show that NEUROG2 is necessary and sufficient to directly transactivate known target genes (NEUROD1, EOMES, RND2). To identify new targets, we engineered NEUROG2-mCherry knock-in human embryonic stem cells for CO generation. The mCherry-high CO cell transcriptome is enriched in extracellular matrix-associated genes, and two genes associated with human-accelerated regions: PPP1R17 and FZD8. We show that NEUROG2 binds COL1A1, COL3A1 and PPP1R17 regulatory elements, and induces their ectopic expression in COs, although NEUROG2 is not required for this expression. Neurog2 similarly induces Col3a1 and Ppp1r17 in murine P19 cells. These data are consistent with a conservation of NEUROG2 function across mammalian species.
Collapse
Affiliation(s)
- Lakshmy Vasan
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Vorapin Chinchalongporn
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Biochemistry, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Fermisk Saleh
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Biochemistry, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Dawn Zinyk
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Biochemistry, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Cao Ke
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Immunology, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Hamsini Suresh
- Department of Physiology, University of Toronto, Medical Sciences Building, 1 King's College Cir, Toronto, ON M5S 1A8, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, 160 College St, Toronto, ON M5S 3E1, Canada
| | - Hussein Ghazale
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Biochemistry, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Lauren Belfiore
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Yacine Touahri
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Biochemistry, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ana-Maria Oproescu
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Shruti Patel
- Department of Medical Biophysics, 101 College St Suite 15-701, Toronto General Hospital, University of Toronto, Toronto, ON M5G 1L7, Canada
- Sunnybrook Research Institute, Physical Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
| | - Matthew Rozak
- Department of Medical Biophysics, 101 College St Suite 15-701, Toronto General Hospital, University of Toronto, Toronto, ON M5G 1L7, Canada
- Sunnybrook Research Institute, Physical Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
| | - Yutaka Amemiya
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sisu Han
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Biochemistry, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Alexandra Moffat
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sandra E. Black
- Dr. Sandra Black Centre for Brain Resilience & Recovery, LC Campbell Cognitive Neurology Unit, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada; Hurvitz Brain Sciences Program
- Department of Medicine (Neurology) (SEB), University of Toronto, Toronto, ON M5S 3H2, Canada
| | - JoAnne McLaurin
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jamie Near
- Department of Medical Biophysics, 101 College St Suite 15-701, Toronto General Hospital, University of Toronto, Toronto, ON M5G 1L7, Canada
- Sunnybrook Research Institute, Physical Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
| | - Arun Seth
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Maged Goubran
- Department of Medical Biophysics, 101 College St Suite 15-701, Toronto General Hospital, University of Toronto, Toronto, ON M5G 1L7, Canada
- Sunnybrook Research Institute, Physical Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
| | - Orly Reiner
- Departments of Molecular Genetics and Molecular Neuroscience, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Jesse Gillis
- Department of Physiology, University of Toronto, Medical Sciences Building, 1 King's College Cir, Toronto, ON M5S 1A8, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, 160 College St, Toronto, ON M5S 3E1, Canada
| | - Chao Wang
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Immunology, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Satoshi Okawa
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Carol Schuurmans
- Sunnybrook Research Institute, Biological Sciences Platform, Hurvitz Brain Sciences Program, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Biochemistry, Medical Sciences Building, 1 King's College Cir, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
2
|
Hatanaka Y, Yamada K, Eritate T, Kawaguchi Y, Hirata T. Neuronal fate resulting from indirect neurogenesis in the mouse neocortex. Cereb Cortex 2024; 34:bhae439. [PMID: 39526524 DOI: 10.1093/cercor/bhae439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/12/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
Excitatory cortical neurons originate from cortical radial glial cells (RGCs). Initially, these neurons were thought to derive directly from RGCs (direct neurogenesis) and be distributed in an inside-out fashion. However, the discovery of indirect neurogenesis, whereby intermediate neuronal progenitors (INPs) generate neurons, challenged this view. To investigate the integration of neurons via these two modes, we developed a method to identify INP progeny and analyze their fate using transgenic mice expressing tamoxifen-inducible Cre recombinase under the neurogenin-2 promoter, alongside thymidine analog incorporation. Their fate was further analyzed using mosaic analysis with double markers in mice. Indirect neurogenesis was prominent during early neurogenesis, generating neuron types that would emerge slightly later than those produced via direct neurogenesis. Despite the timing difference, both neurogenic modes produced fundamentally similar neuron types, as evidenced by marker expression and cortical-depth location. Furthermore, INPs generated pairs of similar phenotype neurons. These findings suggest that indirect neurogenesis, like direct neurogenesis, generates neuron types in a temporally ordered sequence and increases the number of similar neuron types, particularly in deep layers. Thus, both neurogenic modes cooperatively generate a diverse array of neuron types in a similar order, and their progeny populate together to form a coherent cortical structure.
Collapse
Affiliation(s)
- Yumiko Hatanaka
- Laboratory of Cellular and Molecular Neurobiology, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
- Division of Cerebral Circuitry, National Institute for Physiological Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan
- Developmental Neuroscience Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya, Tokyo 156-8506, Japan
| | - Kentaro Yamada
- Laboratory of Cellular and Molecular Neurobiology, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tomoki Eritate
- Laboratory of Cellular and Molecular Neurobiology, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yasuo Kawaguchi
- Division of Cerebral Circuitry, National Institute for Physiological Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan
- Brain Science Institute, Tamagawa University, Machida, Tokyo 194-8610, Japan
| | - Tatsumi Hirata
- Brain Function Laboratory, National Institute of Genetics, SOKENDAI, 1111 Yata, Mishima, Shizuoka 411-8540, Japan
| |
Collapse
|
3
|
Shimojo H, Masaki T, Kageyama R. The Neurog2-Tbr2 axis forms a continuous transition to the neurogenic gene expression state in neural stem cells. Dev Cell 2024; 59:1913-1923.e6. [PMID: 38772376 DOI: 10.1016/j.devcel.2024.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/04/2024] [Accepted: 04/28/2024] [Indexed: 05/23/2024]
Abstract
Neural stem cells (NSCs) differentiate into neuron-fated intermediate progenitor cells (IPCs) via cell division. Although differentiation from NSCs to IPCs is a discrete process, recent transcriptome analyses identified a continuous transcriptional trajectory during this process, raising the question of how to reconcile these contradictory observations. In mouse NSCs, Hes1 expression oscillates, regulating the oscillatory expression of the proneural gene Neurog2, while Hes1 expression disappears in IPCs. Thus, the transition from Hes1 oscillation to suppression is involved in the differentiation of NSCs to IPCs. Here, we found that Neurog2 oscillations induce the accumulation of Tbr2, which suppresses Hes1 expression, generating an IPC-like gene expression state in NSCs. In the absence of Tbr2, Hes1 expression is up-regulated, decreasing the formation of IPCs. These results indicate that the Neurog2-Tbr2 axis forms a continuous transcriptional trajectory to an IPC-like neurogenic state in NSCs, which then differentiate into IPCs via cell division.
Collapse
Affiliation(s)
- Hiromi Shimojo
- Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0871, Japan.
| | - Taimu Masaki
- RIKEN Center for Brain Science, Wako 351-0198, Japan; Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Ryoichiro Kageyama
- RIKEN Center for Brain Science, Wako 351-0198, Japan; Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan; Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
4
|
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
|
5
|
Rosebrock D, Vingron M, Arndt PF. Modeling gene expression cascades during cell state transitions. iScience 2024; 27:109386. [PMID: 38500834 PMCID: PMC10946328 DOI: 10.1016/j.isci.2024.109386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 12/14/2023] [Accepted: 02/27/2024] [Indexed: 03/20/2024] Open
Abstract
During cellular processes such as differentiation or response to external stimuli, cells exhibit dynamic changes in their gene expression profiles. Single-cell RNA sequencing (scRNA-seq) can be used to investigate these dynamic changes. To this end, cells are typically ordered along a pseudotemporal trajectory which recapitulates the progression of cells as they transition from one cell state to another. We infer transcriptional dynamics by modeling the gene expression profiles in pseudotemporally ordered cells using a Bayesian inference approach. This enables ordering genes along transcriptional cascades, estimating differences in the timing of gene expression dynamics, and deducing regulatory gene interactions. Here, we apply this approach to scRNA-seq datasets derived from mouse embryonic forebrain and pancreas samples. This analysis demonstrates the utility of the method to derive the ordering of gene dynamics and regulatory relationships critical for proper cellular differentiation and maturation across a variety of developmental contexts.
Collapse
Affiliation(s)
- Daniel Rosebrock
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Martin Vingron
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Peter F. Arndt
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| |
Collapse
|
6
|
Sokpor G, Kerimoglu C, Ulmke PA, Pham L, Nguyen HD, Brand-Saberi B, Staiger JF, Fischer A, Nguyen HP, Tuoc T. H3 Acetylation-Induced Basal Progenitor Generation and Neocortex Expansion Depends on the Transcription Factor Pax6. BIOLOGY 2024; 13:68. [PMID: 38392287 PMCID: PMC10886678 DOI: 10.3390/biology13020068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 02/24/2024]
Abstract
Enrichment of basal progenitors (BPs) in the developing neocortex is a central driver of cortical enlargement. The transcription factor Pax6 is known as an essential regulator in generation of BPs. H3 lysine 9 acetylation (H3K9ac) has emerged as a crucial epigenetic mechanism that activates the gene expression program required for BP pool amplification. In this current work, we applied immunohistochemistry, RNA sequencing, chromatin immunoprecipitation and sequencing, and the yeast two-hybrid assay to reveal that the BP-genic effect of H3 acetylation is dependent on Pax6 functionality in the developing mouse cortex. In the presence of Pax6, increased H3 acetylation caused BP pool expansion, leading to enhanced neurogenesis, which evoked expansion and quasi-convolution of the mouse neocortex. Interestingly, H3 acetylation activation exacerbates the BP depletion and corticogenesis reduction effect of Pax6 ablation in cortex-specific Pax6 mutants. Furthermore, we found that H3K9 acetyltransferase KAT2A/GCN5 interacts with Pax6 and potentiates Pax6-dependent transcriptional activity. This explains a genome-wide lack of H3K9ac, especially in the promoter regions of BP-genic genes, in the Pax6 mutant cortex. Together, these findings reveal a mechanistic coupling of H3 acetylation and Pax6 in orchestrating BP production and cortical expansion through the promotion of a BP gene expression program during cortical development.
Collapse
Affiliation(s)
- Godwin Sokpor
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
- Lincoln Medical School, University of Lincoln, Lincoln LN6 7TS, UK
| | - Cemil Kerimoglu
- German Center for Neurodegenerative Diseases, 37077 Goettingen, Germany
| | | | - Linh Pham
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Hoang Duy Nguyen
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany
| | - Jochen F Staiger
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
| | - Andre Fischer
- German Center for Neurodegenerative Diseases, 37077 Goettingen, Germany
| | - Huu Phuc Nguyen
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Tran Tuoc
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
| |
Collapse
|
7
|
Péron S, Miyakoshi LM, Brill MS, Manzano-Franco D, Serrano-López J, Fan W, Marichal N, Ghanem A, Conzelmann KK, Karow M, Ortega F, Gascón S, Berninger B. Programming of neural progenitors of the adult subependymal zone towards a glutamatergic neuron lineage by neurogenin 2. Stem Cell Reports 2023; 18:2418-2433. [PMID: 37995703 PMCID: PMC10724369 DOI: 10.1016/j.stemcr.2023.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 11/25/2023] Open
Abstract
Although adult subependymal zone (SEZ) neural stem cells mostly generate GABAergic interneurons, a small progenitor population expresses the proneural gene Neurog2 and produces glutamatergic neurons. Here, we determined whether Neurog2 could respecify SEZ neural stem cells and their progeny toward a glutamatergic fate. Retrovirus-mediated expression of Neurog2 induced the glutamatergic lineage markers TBR2 and TBR1 in cultured SEZ progenitors, which differentiated into functional glutamatergic neurons. Likewise, Neurog2-transduced SEZ progenitors acquired glutamatergic neuron hallmarks in vivo. Intriguingly, they failed to migrate toward the olfactory bulb and instead differentiated within the SEZ or the adjacent striatum, where they received connections from local neurons, as indicated by rabies virus-mediated monosynaptic tracing. In contrast, lentivirus-mediated expression of Neurog2 failed to reprogram early SEZ neurons, which maintained GABAergic identity and migrated to the olfactory bulb. Our data show that NEUROG2 can program SEZ progenitors toward a glutamatergic identity but fails to reprogram their neuronal progeny.
Collapse
Affiliation(s)
- Sophie Péron
- Research Group "Adult Neurogenesis and Cellular Reprogramming", Institute of Physiological Chemistry, University Medical Center Johannes Gutenberg University, Mainz, Germany; Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Leo M Miyakoshi
- Physiological Genomics, Institute of Physiology, Ludwig-Maximilians University Munich, Munich, Germany
| | - Monika S Brill
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Diana Manzano-Franco
- Department of Molecular, Cellular and Developmental Neurobiology, Cajal Institute - CSIC, Madrid, Spain
| | - Julia Serrano-López
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary, Universidad Complutense de Madrid (UCM), Madrid, Spain; Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid, Spain; Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - Wenqiang Fan
- Research Group "Adult Neurogenesis and Cellular Reprogramming", Institute of Physiological Chemistry, University Medical Center Johannes Gutenberg University, Mainz, Germany
| | - Nicolás Marichal
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Alexander Ghanem
- Max von Pettenkofer Institute and Gene Center, Ludwig Maximilians-University Munich, Munich, Germany
| | - Karl-Klaus Conzelmann
- Max von Pettenkofer Institute and Gene Center, Ludwig Maximilians-University Munich, Munich, Germany
| | - Marisa Karow
- Institute of Biochemistry, Friedrich-Alexander Universität Nürnberg-Erlangen, Erlangen, Germany
| | - Felipe Ortega
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary, Universidad Complutense de Madrid (UCM), Madrid, Spain; Instituto Universitario de Investigación en Neuroquímica (IUIN), Madrid, Spain; Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - Sergio Gascón
- Department of Molecular, Cellular and Developmental Neurobiology, Cajal Institute - CSIC, Madrid, Spain.
| | - Benedikt Berninger
- Research Group "Adult Neurogenesis and Cellular Reprogramming", Institute of Physiological Chemistry, University Medical Center Johannes Gutenberg University, Mainz, Germany; Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Focus Program Translational Neurosciences, Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
8
|
Ochi S, Manabe S, Kikkawa T, Osumi N. Thirty Years' History since the Discovery of Pax6: From Central Nervous System Development to Neurodevelopmental Disorders. Int J Mol Sci 2022; 23:6115. [PMID: 35682795 PMCID: PMC9181425 DOI: 10.3390/ijms23116115] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/19/2022] [Accepted: 05/27/2022] [Indexed: 12/23/2022] Open
Abstract
Pax6 is a sequence-specific DNA binding transcription factor that positively and negatively regulates transcription and is expressed in multiple cell types in the developing and adult central nervous system (CNS). As indicated by the morphological and functional abnormalities in spontaneous Pax6 mutant rodents, Pax6 plays pivotal roles in various biological processes in the CNS. At the initial stage of CNS development, Pax6 is responsible for brain patterning along the anteroposterior and dorsoventral axes of the telencephalon. Regarding the anteroposterior axis, Pax6 is expressed inversely to Emx2 and Coup-TF1, and Pax6 mutant mice exhibit a rostral shift, resulting in an alteration of the size of certain cortical areas. Pax6 and its downstream genes play important roles in balancing the proliferation and differentiation of neural stem cells. The Pax6 gene was originally identified in mice and humans 30 years ago via genetic analyses of the eye phenotypes. The human PAX6 gene was discovered in patients who suffer from WAGR syndrome (i.e., Wilms tumor, aniridia, genital ridge defects, mental retardation). Mutations of the human PAX6 gene have also been reported to be associated with autism spectrum disorder (ASD) and intellectual disability. Rodents that lack the Pax6 gene exhibit diverse neural phenotypes, which might lead to a better understanding of human pathology and neurodevelopmental disorders. This review describes the expression and function of Pax6 during brain development, and their implications for neuropathology.
Collapse
Affiliation(s)
| | | | | | - Noriko Osumi
- Department of Developmental Neuroscience, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; (S.O.); (S.M.); (T.K.)
| |
Collapse
|
9
|
Noack F, Vangelisti S, Raffl G, Carido M, Diwakar J, Chong F, Bonev B. Multimodal profiling of the transcriptional regulatory landscape of the developing mouse cortex identifies Neurog2 as a key epigenome remodeler. Nat Neurosci 2022; 25:154-167. [PMID: 35132236 PMCID: PMC8825286 DOI: 10.1038/s41593-021-01002-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 12/14/2021] [Indexed: 12/20/2022]
Abstract
How multiple epigenetic layers and transcription factors (TFs) interact to facilitate brain development is largely unknown. Here, to systematically map the regulatory landscape of neural differentiation in the mouse neocortex, we profiled gene expression and chromatin accessibility in single cells and integrated these data with measurements of enhancer activity, DNA methylation and three-dimensional genome architecture in purified cell populations. This allowed us to identify thousands of new enhancers, their predicted target genes and the temporal relationships between enhancer activation, epigenome remodeling and gene expression. We characterize specific neuronal transcription factors associated with extensive and frequently coordinated changes across multiple epigenetic modalities. In addition, we functionally demonstrate a new role for Neurog2 in directly mediating enhancer activity, DNA demethylation, increasing chromatin accessibility and facilitating chromatin looping in vivo. Our work provides a global view of the gene regulatory logic of lineage specification in the cerebral cortex. By profiling multiple epigenetic layers and enhancer activity in vivo, the authors show a widespread remodeling of the regulatory landscape during mouse cortical development and identify Neurog2 as a key transcription factor driving this process.
Collapse
Affiliation(s)
- Florian Noack
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
| | - Silvia Vangelisti
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
| | - Gerald Raffl
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
| | - Madalena Carido
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
| | - Jeisimhan Diwakar
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
| | - Faye Chong
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
| | - Boyan Bonev
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany. .,Physiological Genomics, Biomedical Center, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
10
|
Bedogni F, Hevner RF. Cell-Type-Specific Gene Expression in Developing Mouse Neocortex: Intermediate Progenitors Implicated in Axon Development. Front Mol Neurosci 2021; 14:686034. [PMID: 34321999 PMCID: PMC8313239 DOI: 10.3389/fnmol.2021.686034] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/03/2021] [Indexed: 01/06/2023] Open
Abstract
Cerebral cortex projection neurons (PNs) are generated from intermediate progenitors (IPs), which are in turn derived from radial glial progenitors (RGPs). To investigate developmental processes in IPs, we profiled IP transcriptomes in embryonic mouse neocortex, using transgenic Tbr2-GFP mice, cell sorting, and microarrays. These data were used in combination with in situ hybridization to ascertain gene sets specific for IPs, RGPs, PNs, interneurons, and other neural and non-neural cell types. RGP-selective transcripts (n = 419) included molecules for Notch receptor signaling, proliferation, neural stem cell identity, apical junctions, necroptosis, hippo pathway, and NF-κB pathway. RGPs also expressed specific genes for critical interactions with meningeal and vascular cells. In contrast, IP-selective genes (n = 136) encoded molecules for activated Delta ligand presentation, epithelial-mesenchymal transition, core planar cell polarity (PCP), axon genesis, and intrinsic excitability. Interestingly, IPs expressed several “dependence receptors” (Unc5d, Dcc, Ntrk3, and Epha4) that induce apoptosis in the absence of ligand, suggesting a competitive mechanism for IPs and new PNs to detect key environmental cues or die. Overall, our results imply a novel role for IPs in the patterning of neuronal polarization, axon differentiation, and intrinsic excitability prior to mitosis. Significantly, IPs highly express Wnt-PCP, netrin, and semaphorin pathway molecules known to regulate axon polarization in other systems. In sum, IPs not only amplify neurogenesis quantitatively, but also molecularly “prime” new PNs for axogenesis, guidance, and excitability.
Collapse
Affiliation(s)
| | - Robert F Hevner
- Department of Pathology, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
11
|
Hoye ML, Silver DL. Decoding mixed messages in the developing cortex: translational regulation of neural progenitor fate. Curr Opin Neurobiol 2021; 66:93-102. [PMID: 33130411 PMCID: PMC8058166 DOI: 10.1016/j.conb.2020.10.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/10/2020] [Accepted: 10/04/2020] [Indexed: 12/16/2022]
Abstract
Regulation of stem cell fate decisions is elemental to faithful development, homeostasis, and organismal fitness. Emerging data demonstrate pluripotent stem cells exhibit a vast transcriptional landscape, which is refined as cells differentiate. In the developing neocortex, transcriptional priming of neural progenitors, coupled with post-transcriptional control, is critical for defining cell fates of projection neurons. In particular, radial glial progenitors exhibit dynamic post-transcriptional regulation, including subcellular mRNA localization, RNA decay, and translation. These processes involve both cis-regulatory and trans-regulatory factors, many of which are implicated in neurodevelopmental disease. This review highlights emerging post-transcriptional mechanisms which govern cortical development, with a particular focus on translational control of neuronal fates, including those relevant for disease.
Collapse
Affiliation(s)
- Mariah L Hoye
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, United States
| | - Debra L Silver
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, United States; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, United States; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, United States; Duke Institute for Brain Sciences, Duke University Medical Center, Durham, NC 27710, United States.
| |
Collapse
|
12
|
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
|
13
|
Hirata T, Shioi G, Abe T, Kiyonari H, Kato S, Kobayashi K, Mori K, Kawasaki T. A Novel Birthdate-Labeling Method Reveals Segregated Parallel Projections of Mitral and External Tufted Cells in the Main Olfactory System. eNeuro 2019; 6:ENEURO.0234-19.2019. [PMID: 31672846 PMCID: PMC6868177 DOI: 10.1523/eneuro.0234-19.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 10/16/2019] [Accepted: 10/19/2019] [Indexed: 01/09/2023] Open
Abstract
A fundamental strategy in sensory coding is parallel processing, whereby unique, distinct features of sensation are computed and projected to the central target in the form of submodal maps. It remains unclear, however, whether such parallel processing strategy is employed in the main olfactory system, which codes the complex hierarchical odor and behavioral scenes. A potential scheme is that distinct subsets of projection neurons in the olfactory bulb (OB) form parallel projections to the targets. Taking advantage of the observation that the distinct projection neurons develop at different times, we developed a Cre-loxP-based method that allows for birthdate-specific labeling of cell bodies and their axon projections in mice. This birthdate tag analysis revealed that the mitral cells (MCs) born in an early developmental stage and the external tufted cells (TCs) born a few days later form segregated parallel projections. Specifically, the latter subset converges the axons onto only two small specific targets, one of which, located at the anterolateral edge of the olfactory tubercle (OT), excludes widespread MC projections. This target is made up of neurons that express dopamine D1 but not D2 receptor and corresponds to the most anterolateral isolation of the CAP compartments (aiCAP) that were defined previously. This finding of segregated projections suggests that olfactory sensing does indeed involve parallel processing of functionally distinct submodalities. Importantly, the birthdate tag method used here may pave the way for deciphering the functional meaning of these individual projection pathways in the future.
Collapse
Affiliation(s)
- Tatsumi Hirata
- Brain Function Laboratory, National Institute of Genetics
- Graduate University for Advanced Studies, SOKENDAI, Mishima 411-8540, Japan
| | - Go Shioi
- Laboratory for Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| | - Takaya Abe
- Laboratory for Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
- Laboratory for Animal Resource Development, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| | - Hiroshi Kiyonari
- Laboratory for Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
- Laboratory for Animal Resource Development, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| | - Shigeki Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Kensaku Mori
- Department of Physiology, Graduate School of Medicine, the University of Tokyo, Tokyo 113-0033, Japan
| | - Takahiko Kawasaki
- Brain Function Laboratory, National Institute of Genetics
- Graduate University for Advanced Studies, SOKENDAI, Mishima 411-8540, Japan
| |
Collapse
|
14
|
Shull G, Haffner C, Huttner WB, Kodandaramaiah SB, Taverna E. Robotic platform for microinjection into single cells in brain tissue. EMBO Rep 2019; 20:e47880. [PMID: 31469223 PMCID: PMC6776899 DOI: 10.15252/embr.201947880] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 07/23/2019] [Accepted: 08/07/2019] [Indexed: 01/02/2023] Open
Abstract
Microinjection into single cells in brain tissue is a powerful technique to study and manipulate neural stem cells. However, such microinjection requires expertise and is a low-throughput process. We developed the "Autoinjector", a robot that utilizes images from a microscope to guide a microinjection needle into tissue to deliver femtoliter volumes of liquids into single cells. The Autoinjector enables microinjection of hundreds of cells within a single organotypic slice, resulting in an overall yield that is an order of magnitude greater than manual microinjection. The Autoinjector successfully targets both apical progenitors (APs) and newborn neurons in the embryonic mouse and human fetal telencephalon. We used the Autoinjector to systematically study gap-junctional communication between neural progenitors in the embryonic mouse telencephalon and found that apical contact is a characteristic feature of the cells that are part of a gap junction-coupled cluster. The throughput and versatility of the Autoinjector will render microinjection an accessible high-performance single-cell manipulation technique and will provide a powerful new platform for performing single-cell analyses in tissue for bioengineering and biophysics applications.
Collapse
Affiliation(s)
- Gabriella Shull
- Department of Biomedical EngineeringUniversity of MinnesotaTwin CitiesMNUSA
- Department of Biomedical EngineeringDuke UniversityDurhamNCUSA
| | - Christiane Haffner
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
| | - Suhasa B Kodandaramaiah
- Department of Biomedical EngineeringUniversity of MinnesotaTwin CitiesMNUSA
- Department of Mechanical EngineeringUniversity of MinnesotaTwin CitiesMNUSA
| | - Elena Taverna
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Max Planck Institute for Evolutionary AnthropologyLeipzigGermany
| |
Collapse
|
15
|
Lzts1 controls both neuronal delamination and outer radial glial-like cell generation during mammalian cerebral development. Nat Commun 2019; 10:2780. [PMID: 31239441 PMCID: PMC6592889 DOI: 10.1038/s41467-019-10730-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 05/24/2019] [Indexed: 01/09/2023] Open
Abstract
In the developing central nervous system, cell departure from the apical surface is the initial and fundamental step to form the 3D, organized architecture. Both delamination of differentiating cells and repositioning of progenitors to generate outer radial glial cells (oRGs) contribute to mammalian neocortical expansion; however, a comprehensive understanding of their mechanisms is lacking. Here, we demonstrate that Lzts1, a molecule associated with microtubule components, promotes both cell departure events. In neuronally committed cells, Lzts1 functions in apical delamination by altering apical junctional organization. In apical RGs (aRGs), Lzts1 expression is variable, depending on Hes1 expression levels. According to its differential levels, Lzts1 induces diverse RG behaviors: planar division, oblique divisions of aRGs that generate oRGs, and their mitotic somal translocation. Loss-of-function of lzts1 impairs all these cell departure processes. Thus, Lzts1 functions as a master modulator of cellular dynamics, contributing to increasing complexity of the cerebral architecture during evolution. Outer radial glial cells (oRGs) are undifferentiated cells that divide in the subventricular zone during neurodevelopment, but the underlying mechanisms are not fully understood. Here the authors show that Lzts1 positively controls both neuronal delamination and generation of oRG-like cell types.
Collapse
|
16
|
Hevner RF. Intermediate progenitors and Tbr2 in cortical development. J Anat 2019; 235:616-625. [PMID: 30677129 DOI: 10.1111/joa.12939] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2019] [Indexed: 12/19/2022] Open
Abstract
In developing cerebral cortex, intermediate progenitors (IPs) are transit amplifying cells that specifically express Tbr2 (gene: Eomes), a T-box transcription factor. IPs are derived from radial glia (RG) progenitors, the neural stem cells of developing cortex. In turn, IPs generate glutamatergic projection neurons (PNs) exclusively. IPs are found in ventricular and subventricular zones, where they differentiate as distinct ventricular IP (vIP) and outer IP (oIP) subtypes. Morphologically, IPs have short processes, resembling filopodia or neurites, that transiently contact other cells, most importantly dividing RG cells to mediate Delta-Notch signaling. Also, IPs secrete a chemokine, Cxcl12, which guides interneuron and microglia migrations and promotes thalamocortical axon growth. In mice, IPs produce clones of 1-12 PNs, sometimes spanning multiple layers. After mitosis, IP daughter cells undergo asymmetric cell death in the majority of instances. In mice, Tbr2 is necessary for PN differentiation and subtype specification, and to repress IP-genic transcription factors. Tbr2 directly represses Insm1, an IP-genic transcription factor gene, as well as Pax6, a key activator of Tbr2 transcription. Without Tbr2, abnormal IPs transiently accumulate in elevated numbers. More broadly, Tbr2 regulates the transcriptome by activating or repressing hundreds of direct target genes. Notably, Tbr2 'unlocks' and activates PN-specific genes, such as Tbr1, by recruiting Jmjd3, a histone H3K27me3 demethylase that removes repressive epigenetic marks placed by polycomb repressive complex 2. IPs have played an important role in the evolution and gyrification of mammalian cerebral cortex, and TBR2 is essential for human brain development.
Collapse
Affiliation(s)
- Robert F Hevner
- Department of Pathology, University of California, San Diego, CA, USA
| |
Collapse
|
17
|
Chromatin Remodeling BAF155 Subunit Regulates the Genesis of Basal Progenitors in Developing Cortex. iScience 2018; 4:109-126. [PMID: 30240734 PMCID: PMC6147019 DOI: 10.1016/j.isci.2018.05.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/22/2018] [Accepted: 05/18/2018] [Indexed: 11/21/2022] Open
Abstract
The abundance of basal progenitors (BPs), basal radial glia progenitors (bRGs) and basal intermediate progenitors (bIPs), in primate brain has been correlated to the high degree of cortical folding. Here we examined the role of BAF155, a subunit of the chromatin remodeling BAF complex, in generation of cortical progenitor heterogeneity. The conditional deletion of BAF155 led to diminished bIP pool and increased number of bRGs, due to delamination of apical RGs. We found that BAF155 is required for normal activity of neurogenic transcription factor PAX6, thus controlling the expression of genes that are involved in bIP specification, cell-cell interaction, and establishment of adherens junction. In a PAX6-dependent manner, BAF155 regulates the expression of the CDC42 effector protein CEP4, thereby controlling progenitor delamination. Furthermore, BAF155-dependent chromatin remodeling seems to exert a specific role in the genesis of BPs through the regulation of human RG-specific genes (such as Foxn4) that possibly acquired evolutionary significance.
Collapse
|
18
|
Mukhtar T, Taylor V. Untangling Cortical Complexity During Development. J Exp Neurosci 2018; 12:1179069518759332. [PMID: 29551911 PMCID: PMC5846925 DOI: 10.1177/1179069518759332] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 01/23/2018] [Indexed: 12/23/2022] Open
Abstract
The cerebral cortex is composed of billions of morphologically and functionally distinct neurons. These neurons are produced and organized in a regimental fashion during development. The ability of neurons to encode and elicit complex cognitive and motor functions depends on their precise molecular processes, identity, and connectivity established during development. Elucidating the cellular and molecular mechanisms that regulate development of the neocortex has been a challenge for many years. The cerebral cortical neuronal subtypes are classified based on morphology, function, intrinsic synaptic properties, location, connectivity, and marker gene expression. Development of the neocortex requires an orchestration of a series of processes including the appropriate determination, migration and positioning of the neurons, acquisition of layer-specific transcriptional hallmarks, and formation of precise axonal projections and networks. Historically, fate mapping, genome-wide analysis, and transcriptome profiling have provided many opportunities for the characterization of neuronal subtypes. During the course of this review, we will address the regimental organization of the cerebral cortex, dissect the cellular subtypes that contribute to cortical complexity, and outline their molecular hallmarks to understand cellular diversity in the cerebral cortex with a focus on the excitatory neurons.
Collapse
Affiliation(s)
- Tanzila Mukhtar
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Verdon Taylor
- Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
19
|
Massimino L, Flores-Garcia L, Di Stefano B, Colasante G, Icoresi-Mazzeo C, Zaghi M, Hamilton BA, Sessa A. TBR2 antagonizes retinoic acid dependent neuronal differentiation by repressing Zfp423 during corticogenesis. Dev Biol 2018; 434:231-248. [PMID: 29305158 PMCID: PMC7032051 DOI: 10.1016/j.ydbio.2017.12.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/26/2017] [Accepted: 12/28/2017] [Indexed: 01/14/2023]
Abstract
During cerebral cortex development, neural progenitors are required to elaborate a variety of cell differentiation signals to which they are continuously exposed. RA acid is a potent inducer of neuronal differentiation as it was found to influence cortical development. We report herein that TBR2, a transcription factor specific to Intermediate (Basal) Neural Progenitors (INPs), represses activation of the RA responsive element and expression of RA target genes in cell lines. This repressive action on RA signaling was functionally confirmed by the decrease of RA-mediated neuronal differentiation in neural stem cells stably overexpressing TBR2. In vivo mapping of RA activity in the developing cortex indicated that RA activity is detected in radial glial cells and subsequently downregulated in INPs, revealing a fine cell-type specific regulation of its signaling. Thus, TBR2 might be a molecular player in opposing RA signaling in INPs. Interestingly, this negative regulation is achieved at least in part by directly repressing the critical nuclear RA co-factor ZFP423. Indeed, we found ZFP423 to be expressed in the developing cortex and promote RA-dependent neuronal differentiation. These data indicate that TBR2 contributes to suppressing RA signaling in INPs, thereby enabling them to re-enter the cell cycle and delay neuronal differentiation.
Collapse
Affiliation(s)
- Luca Massimino
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Lisbeth Flores-Garcia
- Departments of Cellular&Molecular Medicine and Medicine, Moores Cancer Center, and Institute for Genomic Medicine, University of California, 9500 Gilman Drive, La Jolla, San Diego, CA 92093-0644, USA
| | - Bruno Di Stefano
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Gaia Colasante
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Cecilia Icoresi-Mazzeo
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Mattia Zaghi
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Bruce A Hamilton
- Departments of Cellular&Molecular Medicine and Medicine, Moores Cancer Center, and Institute for Genomic Medicine, University of California, 9500 Gilman Drive, La Jolla, San Diego, CA 92093-0644, USA
| | - Alessandro Sessa
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy.
| |
Collapse
|
20
|
Adnani L, Han S, Li S, Mattar P, Schuurmans C. Mechanisms of Cortical Differentiation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 336:223-320. [DOI: 10.1016/bs.ircmb.2017.07.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
21
|
Watanabe Y, Kawaue T, Miyata T. Differentiating cells mechanically limit progenitor cells’ interkinetic nuclear migration to secure apical cytogenesis. Development 2018; 145:dev.162883. [DOI: 10.1242/dev.162883] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Accepted: 06/07/2018] [Indexed: 12/19/2022]
Abstract
Many proliferative epithelia are pseudostratified due to cell cycle–dependent interkinetic nuclear migration (IKNM, basal during G1 and apical during G2). Although most epithelia, including early embryonic neuroepithelia (≤100 µm thick), undergo IKNM over the entire apicobasal extent, more apicobasally elongated (300 µm) neural progenitor cells (also called “radial glia”) in the mid-embryonic mouse cerebral wall move their nuclei only within its apical (100 µm) compartment, leaving the remaining basal part nucleus-free (fiber-like). How this IKNM range (i.e., the thickness of a pseudostratified “ventricular zone” [VZ]) is determined remains unknown. Here, we report external fencing of IKNM and VZ by differentiating cells. When a tight stack of multipolar cells just basal to VZ was “drilled” via acute neuron-directed expression of diphtheria toxin, IKNM of apicobasally connected progenitor cells continued far basally (200 µm). The unfencing-induced, basally overshot nuclei stay in S phase too long and do not move apically, suggesting that external limitation of IKNM is necessary for progenitors to undergo normal cytogenetic behaviors. Thus, physical collaboration between progenitors and differentiating cells including neurons underlies brain development.
Collapse
Affiliation(s)
- Yuto Watanabe
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Takumi Kawaue
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| | - Takaki Miyata
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa, Nagoya 466-8550, Japan
| |
Collapse
|
22
|
Agirman G, Broix L, Nguyen L. Cerebral cortex development: an outside‐in perspective. FEBS Lett 2017; 591:3978-3992. [DOI: 10.1002/1873-3468.12924] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/17/2017] [Accepted: 11/28/2017] [Indexed: 12/18/2022]
Affiliation(s)
- Gulistan Agirman
- GIGA‐Neurosciences Interdisciplinary Cluster for Applied Genoproteomics (GIGA‐R) Liège Belgium
| | - Loïc Broix
- GIGA‐Neurosciences Interdisciplinary Cluster for Applied Genoproteomics (GIGA‐R) Liège Belgium
| | - Laurent Nguyen
- GIGA‐Neurosciences Interdisciplinary Cluster for Applied Genoproteomics (GIGA‐R) Liège Belgium
| |
Collapse
|
23
|
Govindan S, Jabaudon D. Coupling progenitor and neuronal diversity in the developing neocortex. FEBS Lett 2017; 591:3960-3977. [PMID: 28895133 DOI: 10.1002/1873-3468.12846] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/31/2017] [Accepted: 09/06/2017] [Indexed: 12/16/2022]
Abstract
The adult neocortex is composed of several types of glutamatergic neurons, which are sequentially born from progenitors during development. The extent and nature of progenitor diversity, and how it relates to neuronal diversity, is still poorly understood. In this review, we discuss key features of neocortical progenitors across several species, including their morphological properties, cell cycling behaviour and molecular signatures, and how these features relate to the competence of these cells to generate distinct types of progenies.
Collapse
Affiliation(s)
| | - Denis Jabaudon
- Department of Basic Neuroscience, University of Geneva, Switzerland
| |
Collapse
|
24
|
Neurog2 and Ascl1 together regulate a postmitotic derepression circuit to govern laminar fate specification in the murine neocortex. Proc Natl Acad Sci U S A 2017; 114:E4934-E4943. [PMID: 28584103 DOI: 10.1073/pnas.1701495114] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
A derepression mode of cell-fate specification involving the transcriptional repressors Tbr1, Fezf2, Satb2, and Ctip2 operates in neocortical projection neurons to specify six layer identities in sequence. Less well understood is how laminar fate transitions are regulated in cortical progenitors. The proneural genes Neurog2 and Ascl1 cooperate in progenitors to control the temporal switch from neurogenesis to gliogenesis. Here we asked whether these proneural genes also regulate laminar fate transitions. Several defects were observed in the derepression circuit in Neurog2-/-;Ascl1-/- mutants: an inability to repress expression of Tbr1 (a deep layer VI marker) during upper-layer neurogenesis, a loss of Fezf2+/Ctip2+ layer V neurons, and precocious differentiation of normally late-born, Satb2+ layer II-IV neurons. Conversely, in stable gain-of-function transgenics, Neurog2 promoted differentiative divisions and extended the period of Tbr1+/Ctip2+ deep-layer neurogenesis while reducing Satb2+ upper-layer neurogenesis. Similarly, acute misexpression of Neurog2 in early cortical progenitors promoted Tbr1 expression, whereas both Neurog2 and Ascl1 induced Ctip2. However, Neurog2 was unable to influence the derepression circuit when misexpressed in late cortical progenitors, and Ascl1 repressed only Satb2. Nevertheless, neurons derived from late misexpression of Neurog2 and, to a lesser extent, Ascl1, extended aberrant subcortical axon projections characteristic of early-born neurons. Finally, Neurog2 and Ascl1 altered the expression of Ikaros and Foxg1, known temporal regulators. Proneural genes thus act in a context-dependent fashion as early determinants, promoting deep-layer neurogenesis in early cortical progenitors via input into the derepression circuit while also influencing other temporal regulators.
Collapse
|
25
|
Barford K, Yap CC, Dwyer ND, Winckler B. The related neuronal endosomal proteins NEEP21 (Nsg1) and P19 (Nsg2) have divergent expression profiles in vivo. J Comp Neurol 2017; 525:1861-1878. [PMID: 28299779 DOI: 10.1002/cne.24168] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 10/28/2016] [Accepted: 11/18/2016] [Indexed: 01/23/2023]
Abstract
Endosomal maturation and transport constitutes a complex trafficking system present in all cell types. Neurons have adapted their endosomal system to meet their unique and complex needs. These adaptations include repurposing existing proteins to diversify endocytosis and trafficking, as well as preferential expression of certain regulators more highly in neurons than other cell types. These neuronal regulators include the family of Neuron-Specific Gene family members (Nsg), NEEP21 (Nsg1), and P19 (Nsg2). NEEP21/Nsg1 plays a role in the trafficking of multiple receptors, including the cell adhesion molecule L1/NgCAM, the neurotransmitter receptor GluA2, and β-APP. Recently, we showed that NEEP2/Nsg1 and P19/Nsg2 are not expressed in all neuronal cell types in vitro. However, it is not known where and when NEEP21/Nsg1 and P19/Nsg2 are expressed in vivo, and whether both proteins are always coexpressed. Here, we show that NEEP21/Nsg1 and P19/Nsg2 are present in both overlapping and distinct cell populations in the hippocampus, neocortex, and cerebellum during development. NEEP21/Nsg1 and P19/Nsg2 levels are highest during embryonic development, and expression persists in the juvenile mouse brain. In particular, a subset of layer V cortical neurons retains relatively high expression of both NEEP21/Nsg1 and P19/Nsg2 at postnatal day 16 as well as in the CA1-3 regions of the hippocampus. In the cerebellum, NEEP21/Nsg1 expression becomes largely restricted to Purkinje neurons in adulthood whereas P19/Nsg2 expression strikingly disappears from the cerebellum with age. This divergent and restricted expression likely reflects differential needs for this class of trafficking regulators in different neurons during different stages of maturation.
Collapse
Affiliation(s)
- Kelly Barford
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| | - Chan Choo Yap
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| | - Noelle D Dwyer
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| | - Bettina Winckler
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
26
|
Marinaro F, Marzi MJ, Hoffmann N, Amin H, Pelizzoli R, Niola F, Nicassio F, De Pietri Tonelli D. MicroRNA-independent functions of DGCR8 are essential for neocortical development and TBR1 expression. EMBO Rep 2017; 18:603-618. [PMID: 28232627 DOI: 10.15252/embr.201642800] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 01/16/2017] [Accepted: 01/19/2017] [Indexed: 12/31/2022] Open
Abstract
Recent evidence indicates that the miRNA biogenesis factors DROSHA, DGCR8, and DICER exert non-overlapping functions, and have also roles in miRNA-independent regulatory mechanisms. However, it is currently unknown whether miRNA-independent functions of DGCR8 play any role in the maintenance of neuronal progenitors and during corticogenesis. Here, by phenotypic comparison of cortices from conditional Dgcr8 and Dicer knockout mice, we show that Dgcr8 deletion, in contrast to Dicer depletion, leads to premature differentiation of neural progenitor cells and overproduction of TBR1-positive neurons. Remarkably, depletion of miRNAs upon DCGR8 loss is reduced compared to DICER loss, indicating that these phenotypic differences are mediated by miRNA-independent functions of DGCR8. We show that Dgcr8 mutations induce an earlier and stronger phenotype in the developing nervous system compared to Dicer mutants and that miRNA-independent functions of DGCR8 are critical for corticogenesis. Finally, our data also suggest that the Microprocessor complex, with DROSHA and DGCR8 as core components, directly regulates the Tbr1 transcript, containing evolutionarily conserved hairpins that resemble miRNA precursors, independently of miRNAs.
Collapse
Affiliation(s)
- Federica Marinaro
- Neuroscience and Brain Technologies Department, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Matteo J Marzi
- Center for Genomic Science of IIT@SEMM, Istituto Italiano di Tecnologia, Milan, Italy
| | - Nadin Hoffmann
- Neuroscience and Brain Technologies Department, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Hayder Amin
- Neuroscience and Brain Technologies Department, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Roberta Pelizzoli
- Neuroscience and Brain Technologies Department, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Francesco Niola
- Neuroscience and Brain Technologies Department, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Francesco Nicassio
- Center for Genomic Science of IIT@SEMM, Istituto Italiano di Tecnologia, Milan, Italy
| | | |
Collapse
|
27
|
Cipriani S, Journiac N, Nardelli J, Verney C, Delezoide AL, Guimiot F, Gressens P, Adle-Biassette H. Dynamic Expression Patterns of Progenitor and Neuron Layer Markers in the Developing Human Dentate Gyrus and Fimbria. Cereb Cortex 2017; 27:358-372. [PMID: 26443441 PMCID: PMC5894254 DOI: 10.1093/cercor/bhv223] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The molecular mechanisms that orchestrate the development of the human dentate gyrus are not known. In this study, we characterized the formation of human dentate and fimbrial progenitors and postmitotic neurons from 9 gestational weeks (GW9) to GW25. PAX6+ progenitor cells remained proliferative until GW16 in the dentate ventricular zone. By GW11, the secondary dentate matrix had developed in the intermediate zone, surrounding the dentate anlage and streaming toward the subpial layer. This secondary matrix contained proliferating PAX6+ and/or TBR2+ progenitors. In parallel, SOX2+ and PAX6+ fimbrial cells were detected approaching the dentate anlage, representing a possible source of extra-dentate progenitors. By GW16, when the granule cell layer could be delineated, a hilar matrix containing PAX6+ and some TBR2+ progenitors had become identifiable. By GW25, when the 2 limbs of the granule cell layer had formed, the secondary dentate matrix was reduced to a pool of progenitors at the fimbrio-dentate junction. Although human dentate development recapitulates key steps previously described in rodents, differences seemed to emerge in neuron layer markers expression. Further studies are necessary to better elucidate their role in dentate formation and connectivity.
Collapse
Affiliation(s)
- Sara Cipriani
- INSERM UMR 1141, Hôpital Robert-Debré, Paris, France
- Faculté de Médecine Denis Diderot, Université Paris 7, Paris, France
| | - Nathalie Journiac
- INSERM UMR 1141, Hôpital Robert-Debré, Paris, France
- Faculté de Médecine Denis Diderot, Université Paris 7, Paris, France
| | - Jeannette Nardelli
- INSERM UMR 1141, Hôpital Robert-Debré, Paris, France
- Faculté de Médecine Denis Diderot, Université Paris 7, Paris, France
| | - Catherine Verney
- INSERM UMR 1141, Hôpital Robert-Debré, Paris, France
- Faculté de Médecine Denis Diderot, Université Paris 7, Paris, France
| | - Anne-Lise Delezoide
- INSERM UMR 1141, Hôpital Robert-Debré, Paris, France
- Faculté de Médecine Denis Diderot, Université Paris 7, Paris, France
- Service de Biologie du Développement, Hôpital Robert-Debré, APHP, Paris, France
| | - Fabien Guimiot
- INSERM UMR 1141, Hôpital Robert-Debré, Paris, France
- Faculté de Médecine Denis Diderot, Université Paris 7, Paris, France
- Service de Biologie du Développement, Hôpital Robert-Debré, APHP, Paris, France
| | - Pierre Gressens
- INSERM UMR 1141, Hôpital Robert-Debré, Paris, France
- Faculté de Médecine Denis Diderot, Université Paris 7, Paris, France
| | - Homa Adle-Biassette
- INSERM UMR 1141, Hôpital Robert-Debré, Paris, France
- Faculté de Médecine Denis Diderot, Université Paris 7, Paris, France
- Service d'Anatomie et de Cytologie Pathologiques, Hôpital Lariboisère, APHP, Paris, France
| |
Collapse
|
28
|
Nagasaka A, Shinoda T, Kawaue T, Suzuki M, Nagayama K, Matsumoto T, Ueno N, Kawaguchi A, Miyata T. Differences in the Mechanical Properties of the Developing Cerebral Cortical Proliferative Zone between Mice and Ferrets at both the Tissue and Single-Cell Levels. Front Cell Dev Biol 2016; 4:139. [PMID: 27933293 PMCID: PMC5122735 DOI: 10.3389/fcell.2016.00139] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 11/11/2016] [Indexed: 11/13/2022] Open
Abstract
Cell-producing events in developing tissues are mechanically dynamic throughout the cell cycle. In many epithelial systems, cells are apicobasally tall, with nuclei and somata that adopt different apicobasal positions because nuclei and somata move in a cell cycle-dependent manner. This movement is apical during G2 phase and basal during G1 phase, whereas mitosis occurs at the apical surface. These movements are collectively referred to as interkinetic nuclear migration, and such epithelia are called "pseudostratified." The embryonic mammalian cerebral cortical neuroepithelium is a good model for highly pseudostratified epithelia, and we previously found differences between mice and ferrets in both horizontal cellular density (greater in ferrets) and nuclear/somal movements (slower during G2 and faster during G1 in ferrets). These differences suggest that neuroepithelial cells alter their nucleokinetic behavior in response to physical factors that they encounter, which may form the basis for evolutionary transitions toward more abundant brain-cell production from mice to ferrets and primates. To address how mouse and ferret neuroepithelia may differ physically in a quantitative manner, we used atomic force microscopy to determine that the vertical stiffness of their apical surface is greater in ferrets (Young's modulus = 1700 Pa) than in mice (1400 Pa). We systematically analyzed factors underlying the apical-surface stiffness through experiments to pharmacologically inhibit actomyosin or microtubules and to examine recoiling behaviors of the apical surface upon laser ablation and also through electron microscopy to observe adherens junction. We found that although both actomyosin and microtubules are partly responsible for the apical-surface stiffness, the mouse<ferret relationship in the apical-surface stiffness was maintained even in the presence of inhibitors. We also found that the stiffness of single, dissociated neuroepithelial cells is actually greater in mice (720 Pa) than in ferrets (450 Pa). Adherens junction was ultrastructurally comparable between mice and ferrets. These results show that the horizontally denser packing of neuroepithelial cell processes is a major contributor to the increased tissue-level apical stiffness in ferrets, and suggest that tissue-level mechanical properties may be achieved by balancing cellular densification and the physical properties of single cells.
Collapse
Affiliation(s)
- Arata Nagasaka
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Nagoya University Nagoya, Japan
| | - Tomoyasu Shinoda
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Nagoya University Nagoya, Japan
| | - Takumi Kawaue
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Nagoya University Nagoya, Japan
| | - Makoto Suzuki
- Division for Morphogenesis, Department of Developmental Biology, National Institute for Basic Biology Okazaki, Japan
| | - Kazuaki Nagayama
- Micro-Nano Biomechanics Laboratory, Department of Intelligent Systems Engineering, Ibaraki University Hitachi, Japan
| | - Takeo Matsumoto
- Biomechanics Laboratory, Department of Mechanical Engineering, Nagoya Institute of Technology Nagoya, Japan
| | - Naoto Ueno
- Division for Morphogenesis, Department of Developmental Biology, National Institute for Basic Biology Okazaki, Japan
| | - Ayano Kawaguchi
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Nagoya University Nagoya, Japan
| | - Takaki Miyata
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Nagoya University Nagoya, Japan
| |
Collapse
|
29
|
Abstract
T-box transcription factors play key roles in the regulation of developmental processes such as cell differentiation and migration. Mammals have 17 T-box genes, of which several regulate brain development. The Tbr1 subfamily of T-box genes is particularly important in development of the cerebral cortex, olfactory bulbs (OBs), and cerebellum. This subfamily is comprised of Tbr1, Tbr2 (also known as Eomes), and Tbx21. In developing cerebral cortex, Tbr2 and Tbr1 are expressed during successive stages of differentiation in the pyramidal neuron lineage, from Tbr2+ intermediate progenitors to Tbr1+ postmitotic glutamatergic neurons. At each stage, Tbr2 and Tbr1 regulate laminar and regional identity of cortical projection neurons, cell migration, and axon guidance. In the OB, Tbr1 subfamily genes regulate neurogenesis of mitral and tufted cells, and glutamatergic juxtaglomerular interneurons. Tbr2 is also prominent in the development of retinal ganglion cells in nonimage-forming pathways. Other regions that require Tbr2 or Tbr1 in development or adulthood include the cerebellum and adult dentate gyrus. In humans, de novo mutations in TBR1 are important causes of sporadic autism and intellectual disability. Further studies of T-box transcription factors will enhance our understanding of neurodevelopmental disorders and inform approaches to new therapies.
Collapse
|
30
|
Orchestration of Neuronal Differentiation and Progenitor Pool Expansion in the Developing Cortex by SoxC Genes. J Neurosci 2015. [PMID: 26203155 DOI: 10.1523/jneurosci.1663-15.2015] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
As the cerebral cortex forms, specialized molecular cascades direct the expansion of progenitor pools, the differentiation of neurons, or the maturation of discrete neuronal subtypes, together ensuring that the correct amounts and classes of neurons are generated. In several neural systems, the SoxC transcriptional regulators, particularly Sox11 and Sox4, have been characterized as functioning exclusively and redundantly in promoting neuronal differentiation. Using the mouse cerebral cortex as a model, Sox11 and Sox4 were examined in the formation of the most complex part of the mammalian brain. Anticipated prodifferentiation roles were observed. Distinct expression patterns and mutant phenotypes, however, reveal that Sox11 and Sox4 are not redundant in the cortex, but rather act in overlapping and discrete populations of neurons. In particular, Sox11 acts in early-born neurons; binding to its partner protein, Neurogenin1, leads to selective targeting and transactivation of a downstream gene, NeuroD1. In addition to neuronal expression, Sox4 was unexpectedly expressed in intermediate progenitor cells, the transit amplifying cell of the cerebral cortex. Sox4 mutant analyses reveal a requirement for Sox4 in IPC specification and maintenance. In intermediate progenitors, Sox4 partners with the proneural gene Neurogenin2 to activate Tbrain2 and then with Tbrain2 to maintain this cell fate. This work reveals an intricately structured molecular architecture for SoxC molecules, with Sox11 acting in a select set of cortical neurons and Sox4 playing an unanticipated role in designating secondary progenitors.
Collapse
|
31
|
Cipriani S, Nardelli J, Verney C, Delezoide AL, Guimiot F, Gressens P, Adle-Biassette H. Dynamic Expression Patterns of Progenitor and Pyramidal Neuron Layer Markers in the Developing Human Hippocampus. Cereb Cortex 2015; 26:1255-71. [DOI: 10.1093/cercor/bhv079] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
32
|
Signal transducer and activator of transcription-3 maintains the stemness of radial glia at mid-neurogenesis. J Neurosci 2015; 35:1011-23. [PMID: 25609618 DOI: 10.1523/jneurosci.2119-14.2015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Radial glial cells are stem cell-like populations of glial nature that supply neurons either directly or indirectly via basal progenitors that give rise to neurons. Here we show that signal transducer and activator of transcription-3 (STAT3) signaling, a cytokine signaling mediated by Janus tyrosine kinase (Jak), is active during neurogenesis in radial glia (RG) but not in basal progenitors. Enhanced STAT3 signaling in cortical progenitors caused more RG to persist rather than become neurons. Targeted deletion or RNAi-mediated knockdown of Stat3 resulted in fewer radial glial cells and more basal progenitors and led to premature neurogenesis. The neuronal populations affected in Stat3 mutant mice were the late-born neurons that constitute the upper cortical layers rather than early-born neurons, thus supporting the view that the role of STAT3 at mid-neurogenesis is layer specific. Analysis of dividing RG revealed that STAT3 selectively increased the proportion of dividing RG, whereas downregulation of STAT3 reduced the proportion. Consistent with this, STAT3 activity in dividing RG was associated frequently with vertical cleavage. Pair-cell analysis showed that elevated STAT3 activity correlated with symmetric division of RG, producing more RG, whereas elimination of STAT3 generated more neurogenic cells. Together, our results suggest that STAT3 maintains the stemness of RG and inhibits their transition to basal progenitors at mid-neurogenesis, so probably preserving a pool of RG for later neurogenesis or gliogenesis.
Collapse
|
33
|
Johnson MB, Wang PP, Atabay KD, Murphy EA, Doan RN, Hecht JL, Walsh CA. Single-cell analysis reveals transcriptional heterogeneity of neural progenitors in human cortex. Nat Neurosci 2015; 18:637-46. [PMID: 25734491 DOI: 10.1038/nn.3980] [Citation(s) in RCA: 213] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 02/24/2015] [Indexed: 12/20/2022]
Abstract
The human cerebral cortex depends for its normal development and size on a precisely controlled balance between self-renewal and differentiation of diverse neural progenitor cells. Specialized progenitors that are common in humans but virtually absent in rodents, called outer radial glia (ORG), have been suggested to be crucial to the evolutionary expansion of the human cortex. We combined progenitor subtype-specific sorting with transcriptome-wide RNA sequencing to identify genes enriched in human ORG, which included targets of the transcription factor neurogenin and previously uncharacterized, evolutionarily dynamic long noncoding RNAs. Activating the neurogenin pathway in ferret progenitors promoted delamination and outward migration. Finally, single-cell transcriptional profiling in human, ferret and mouse revealed more cells coexpressing proneural neurogenin targets in human than in other species, suggesting greater neuronal lineage commitment and differentiation of self-renewing progenitors. Thus, we find that the abundance of human ORG is paralleled by increased transcriptional heterogeneity of cortical progenitors.
Collapse
Affiliation(s)
- Matthew B Johnson
- 1] Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts, USA. [2] Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts, USA. [3] Howard Hughes Medical Institute, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Peter P Wang
- 1] Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts, USA. [2] Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts, USA. [3] Howard Hughes Medical Institute, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Kutay D Atabay
- 1] Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts, USA. [2] Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts, USA. [3] Howard Hughes Medical Institute, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Elisabeth A Murphy
- 1] Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts, USA. [2] Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts, USA. [3] Howard Hughes Medical Institute, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Ryan N Doan
- 1] Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts, USA. [2] Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts, USA. [3] Howard Hughes Medical Institute, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Jonathan L Hecht
- 1] Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA. [2] Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| | - Christopher A Walsh
- 1] Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts, USA. [2] Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, Massachusetts, USA. [3] Howard Hughes Medical Institute, Boston Children's Hospital, Boston, Massachusetts, USA. [4] Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA. [5] Department of Neurology, Harvard Medical School, Boston, Massachusetts, USA. [6] Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
34
|
Artegiani B, de Jesus Domingues AM, Bragado Alonso S, Brandl E, Massalini S, Dahl A, Calegari F. Tox: a multifunctional transcription factor and novel regulator of mammalian corticogenesis. EMBO J 2014; 34:896-910. [PMID: 25527292 DOI: 10.15252/embj.201490061] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 12/03/2014] [Indexed: 01/17/2023] Open
Abstract
Major efforts are invested to characterize the factors controlling the proliferation of neural stem cells. During mammalian corticogenesis, our group has identified a small pool of genes that are transiently downregulated in the switch of neural stem cells to neurogenic division and reinduced in newborn neurons. Among these switch genes, we found Tox, a transcription factor with hitherto uncharacterized roles in the nervous system. Here, we investigated the role of Tox in corticogenesis by characterizing its expression at the tissue, cellular and temporal level. We found that Tox is regulated by calcineurin/Nfat signalling. Moreover, we combined DNA adenine methyltransferase identification (DamID) with deep sequencing to characterize the chromatin binding properties of Tox including its motif and downstream transcriptional targets including Sox2, Tbr2, Prox1 and other key factors. Finally, we manipulated Tox in the developing brain and validated its multiple roles in promoting neural stem cell proliferation and neurite outgrowth of newborn neurons. Our data provide a valuable resource to study the role of Tox in other tissues and highlight a novel key player in brain development.
Collapse
Affiliation(s)
- Benedetta Artegiani
- DFG-Research Center for Regenerative Therapies, Cluster of Excellence, TU-Dresden, Dresden, Germany
| | | | - Sara Bragado Alonso
- DFG-Research Center for Regenerative Therapies, Cluster of Excellence, TU-Dresden, Dresden, Germany
| | - Elisabeth Brandl
- DFG-Research Center for Regenerative Therapies, Cluster of Excellence, TU-Dresden, Dresden, Germany
| | - Simone Massalini
- DFG-Research Center for Regenerative Therapies, Cluster of Excellence, TU-Dresden, Dresden, Germany
| | - Andreas Dahl
- Deep Sequencing Group-SFB655, Biotechnology Center, TU-Dresden, Dresden, Germany
| | - Federico Calegari
- DFG-Research Center for Regenerative Therapies, Cluster of Excellence, TU-Dresden, Dresden, Germany
| |
Collapse
|
35
|
Progenitor genealogy in the developing cerebral cortex. Cell Tissue Res 2014; 359:17-32. [PMID: 25141969 DOI: 10.1007/s00441-014-1979-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 07/28/2014] [Indexed: 10/24/2022]
Abstract
The mammalian cerebral cortex is characterized by a complex histological organization that reflects the spatio-temporal stratifications of related stem and neural progenitor cells, which are responsible for the generation of distinct glial and neuronal subtypes during development. Some work has been done to shed light on the existing filiations between these progenitors as well as their respective contribution to cortical neurogenesis. The aim of the present review is to summarize the current views of progenitor hierarchy and relationship in the developing cortex and to further discuss future research directions that would help us to understand the molecular and cellular regulating mechanisms involved in cerebral corticogenesis.
Collapse
|
36
|
Vied CM, Freudenberg F, Wang Y, Raposo AASF, Feng D, Nowakowski RS. A multi-resource data integration approach: identification of candidate genes regulating cell proliferation during neocortical development. Front Neurosci 2014; 8:257. [PMID: 25191221 PMCID: PMC4139594 DOI: 10.3389/fnins.2014.00257] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 08/01/2014] [Indexed: 11/17/2022] Open
Abstract
Neurons of the mammalian neocortex are produced by proliferating cells located in the ventricular zone (VZ) lining the lateral ventricles. This is a complex and sequential process, requiring precise control of cell cycle progression, fate commitment and differentiation. We have analyzed publicly available databases from mouse and human to identify candidate genes that are potentially involved in regulating early neocortical development and neurogenesis. We used a mouse in situ hybridization dataset (The Allen Institute for Brain Science) to identify 13 genes (Cdon, Celsr1, Dbi, E2f5, Eomes, Hmgn2, Neurog2, Notch1, Pcnt, Sox3, Ssrp1, Tead2, Tgif2) with high correlation of expression in the proliferating cells of the VZ of the neocortex at early stages of development (E15.5). We generated a similar human brain network using microarray and RNA-seq data (BrainSpan Atlas) and identified 407 genes with high expression in the developing human VZ and subventricular zone (SVZ) at 8–9 post-conception weeks. Seven of the human genes were also present in the mouse VZ network. The human and mouse networks were extended using available genetic and proteomic datasets through GeneMANIA. A gene ontology search of the mouse and human networks indicated that many of the genes are involved in the cell cycle, DNA replication, mitosis and transcriptional regulation. The reported involvement of Cdon, Celsr1, Dbi, Eomes, Neurog2, Notch1, Pcnt, Sox3, Tead2, and Tgif2 in neural development or diseases resulting from the disruption of neurogenesis validates these candidate genes. Taken together, our knowledge-based discovery method has validated the involvement of many genes already known to be involved in neocortical development and extended the potential number of genes by 100's, many of which are involved in functions related to cell proliferation but others of which are potential candidates for involvement in the regulation of neocortical development.
Collapse
Affiliation(s)
- Cynthia M Vied
- Department of Biomedical Sciences, College of Medicine, Florida State University Tallahassee, FL, USA
| | - Florian Freudenberg
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Frankfurt Frankfurt, Germany
| | - Yuting Wang
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore Singapore, Singapore
| | | | - David Feng
- Allen Institute for Brain Science Seattle, WA, USA
| | - Richard S Nowakowski
- Department of Biomedical Sciences, College of Medicine, Florida State University Tallahassee, FL, USA
| |
Collapse
|
37
|
Kawaue T, Sagou K, Kiyonari H, Ota K, Okamoto M, Shinoda T, Kawaguchi A, Miyata T. Neurogenin2-d4Venus and Gadd45g-d4Venus transgenic mice: visualizing mitotic and migratory behaviors of cells committed to the neuronal lineage in the developing mammalian brain. Dev Growth Differ 2014; 56:293-304. [PMID: 24712911 PMCID: PMC4477914 DOI: 10.1111/dgd.12131] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 03/08/2014] [Accepted: 03/08/2014] [Indexed: 12/22/2022]
Abstract
To achieve highly sensitive and comprehensive assessment of the morphology and dynamics of cells committed to the neuronal lineage in mammalian brain primordia, we generated two transgenic mouse lines expressing a destabilized (d4) Venus controlled by regulatory elements of the Neurogenin2 (Neurog2) or Gadd45g gene. In mid-embryonic neocortical walls, expression of Neurog2-d4Venus mostly overlapped with that of Neurog2 protein, with a slightly (1 h) delayed onset. Although Neurog2-d4Venus and Gadd45g-d4Venus mice exhibited very similar labeling patterns in the ventricular zone (VZ), in Gadd45g-d4Venus mice cells could be visualized in more basal areas containing fully differentiated neurons, where Neurog2-d4Venus fluorescence was absent. Time-lapse monitoring revealed that most d4Venus+ cells in the VZ had processes extending to the apical surface; many of these cells eventually retracted their apical process and migrated basally to the subventricular zone, where neurons, as well as the intermediate neurogenic progenitors that undergo terminal neuron-producing division, could be live-monitored by d4Venus fluorescence. Some d4Venus+ VZ cells instead underwent nuclear migration to the apical surface, where they divided to generate two d4Venus+ daughter cells, suggesting that the symmetric terminal division that gives rise to neuron pairs at the apical surface can be reliably live-monitored. Similar lineage-committed cells were observed in other developing neural regions including retina, spinal cord, and cerebellum, as well as in regions of the peripheral nervous system such as dorsal root ganglia. These mouse lines will be useful for elucidating the cellular and molecular mechanisms underlying development of the mammalian nervous system.
Collapse
Affiliation(s)
- Takumi Kawaue
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, 65 Tsurumai, Showa-ku, Nagoya, 466-8550, Japan
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Imayoshi I, Kageyama R. bHLH Factors in Self-Renewal, Multipotency, and Fate Choice of Neural Progenitor Cells. Neuron 2014; 82:9-23. [DOI: 10.1016/j.neuron.2014.03.018] [Citation(s) in RCA: 203] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2014] [Indexed: 12/18/2022]
|
39
|
Paridaen JTML, Huttner WB. Neurogenesis during development of the vertebrate central nervous system. EMBO Rep 2014; 15:351-64. [PMID: 24639559 DOI: 10.1002/embr.201438447] [Citation(s) in RCA: 261] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
During vertebrate development, a wide variety of cell types and tissues emerge from a single fertilized oocyte. One of these tissues, the central nervous system, contains many types of neurons and glial cells that were born during the period of embryonic and post-natal neuro- and gliogenesis. As to neurogenesis, neural progenitors initially divide symmetrically to expand their pool and switch to asymmetric neurogenic divisions at the onset of neurogenesis. This process involves various mechanisms involving intrinsic as well as extrinsic factors. Here, we discuss the recent advances and insights into regulation of neurogenesis in the developing vertebrate central nervous system. Topics include mechanisms of (a)symmetric cell division, transcriptional and epigenetic regulation, and signaling pathways, using mostly examples from the developing mammalian neocortex.
Collapse
|
40
|
Abstract
Proneural genes encode evolutionarily conserved basic-helix-loop-helix transcription factors. In Drosophila, proneural genes are required and sufficient to confer a neural identity onto naïve ectodermal cells, inducing delamination and subsequent neuronal differentiation. In vertebrates, proneural genes are expressed in cells that already have a neural identity, but they are still required and sufficient to initiate neurogenesis. In all organisms, proneural genes control neurogenesis by regulating Notch-mediated lateral inhibition and initiating the expression of downstream differentiation genes. The general mode of proneural gene function has thus been elucidated. However, the regulatory mechanisms that spatially and temporally control proneural gene function are only beginning to be deciphered. Understanding how proneural gene function is regulated is essential, as aberrant proneural gene expression has recently been linked to a variety of human diseases-ranging from cancer to neuropsychiatric illnesses and diabetes. Recent insights into proneural gene function in development and disease are highlighted herein.
Collapse
Affiliation(s)
- Carol Huang
- Department of Pediatrics, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Jennifer A Chan
- Department of Pathology & Laboratory Medicine, Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada.
| | - Carol Schuurmans
- Department of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
41
|
Cain JT, Berosik MA, Snyder SD, Crawford NF, Nour SI, Schaubhut GJ, Darland DC. Shifts in the vascular endothelial growth factor isoforms result in transcriptome changes correlated with early neural stem cell proliferation and differentiation in mouse forebrain. Dev Neurobiol 2013; 74:63-81. [PMID: 24124161 DOI: 10.1002/dneu.22130] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 08/21/2013] [Accepted: 09/04/2013] [Indexed: 12/12/2022]
Abstract
Regulation of neural stem cell (NSC) fate decisions is critical during the transition from a multicellular mammalian forebrain neuroepithelium to the multilayered neocortex. Forebrain development requires coordinated vascular investment alongside NSC differentiation. Vascular endothelial growth factor A (Vegf) has proven to be a pleiotrophic gene whose multiple protein isoforms regulate a broad range of effects in neurovascular systems. To test the hypothesis that the Vegf isoforms (120, 164, and 188) are required for normal forebrain development, we analyzed the forebrain transcriptome of mice expressing specific Vegf isoforms, Vegf120, VegfF188, or a combination of Vegf120/188. Transcriptome analysis identified differentially expressed genes in embryonic day (E) 9.5 forebrain, a time point preceding dramatic neuroepithelial expansion and vascular investment in the telencephalon. Meta-analysis identified gene pathways linked to chromosome-level modifications, cell fate regulation, and neurogenesis that were altered in Vegf isoform mice. Based on these gene network shifts, we predicted that NSC populations would be affected in later stages of forebrain development. In the E11.5 telencephalon, we quantified mitotic cells [Phospho-Histone H3 (pHH3)-positive] and intermediate progenitor cells (Tbr2/Eomes-positive), observing quantitative and qualitative shifts in these populations. We observed qualitative shifts in cortical layering at P0, particularly with Ctip2-positive cells in layer V. The results identify a suite of genes and functional gene networks that can be used to further dissect the role of Vegf in regulating NSC differentiation and downstream consequences for NSC fate decisions.
Collapse
Affiliation(s)
- Jacob T Cain
- Department of Biology, University of North Dakota, Grand Forks, North Dakota
| | | | | | | | | | | | | |
Collapse
|
42
|
Prenatal exposure to suberoylanilide hydroxamic acid perturbs corticogenesis. Neurosci Res 2013; 77:42-9. [DOI: 10.1016/j.neures.2013.06.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 06/14/2013] [Accepted: 06/26/2013] [Indexed: 11/30/2022]
|
43
|
Wilkinson G, Dennis D, Schuurmans C. Proneural genes in neocortical development. Neuroscience 2013; 253:256-73. [PMID: 23999125 DOI: 10.1016/j.neuroscience.2013.08.029] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 08/16/2013] [Accepted: 08/18/2013] [Indexed: 02/01/2023]
Abstract
Neurons, astrocytes and oligodendrocytes arise from CNS progenitor cells at defined times and locations during development, with transcription factors serving as key determinants of these different neural cell fates. An emerging theme is that the transcription factors that specify CNS cell fates function in a context-dependent manner, regulated by post-translational modifications and epigenetic alterations that partition the genome (and hence target genes) into active or silent domains. Here we profile the critical roles of the proneural genes, which encode basic-helix-loop-helix (bHLH) transcription factors, in specifying neural cell identities in the developing neocortex. In particular, we focus on the proneural genes Neurogenin 1 (Neurog1), Neurog2 and Achaete scute-like 1 (Ascl1), which are each expressed in a distinct fashion in the progenitor cell pools that give rise to all of the neuronal and glial cell types of the mature neocortex. Notably, while the basic functions of these proneural genes have been elucidated, it is becoming increasingly evident that tight regulatory controls dictate when, where and how they function. Current efforts to better understand how proneural gene function is regulated will not only improve our understanding of neocortical development, but are also critical to the future development of regenerative therapies for the treatment of neuronal degeneration or disease.
Collapse
Affiliation(s)
- G Wilkinson
- Hotchkiss Brain Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | | | | |
Collapse
|
44
|
Itoh Y, Tyssowski K, Gotoh Y. Transcriptional coupling of neuronal fate commitment and the onset of migration. Curr Opin Neurobiol 2013; 23:957-64. [PMID: 23973158 DOI: 10.1016/j.conb.2013.08.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Revised: 08/05/2013] [Accepted: 08/05/2013] [Indexed: 12/18/2022]
Abstract
During mammalian CNS development, when the neural precursor cells commit to the neuronal fate they must delaminate and migrate toward the pial surface in order to reach the appropriate final location. Thus, the coordination of delamination and fate commitment is important in creating the correct structure. Although previous studies have proposed that spindle orientation during mitosis plays a role in both delamination and fate commitment, thus coordinating these events, subsequent studies have challenged this model. Recent work has identified several transcriptional mechanisms associated with neurogenesis that inhibit cell adhesion of newborn neurons and intermediate neuronal progenitors, thereby triggering delamination and linking it with fate commitment.
Collapse
Affiliation(s)
- Yasuhiro Itoh
- Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan.
| | | | | |
Collapse
|
45
|
Nelson BR, Hodge RD, Bedogni F, Hevner RF. Dynamic interactions between intermediate neurogenic progenitors and radial glia in embryonic mouse neocortex: potential role in Dll1-Notch signaling. J Neurosci 2013; 33:9122-39. [PMID: 23699523 PMCID: PMC3716275 DOI: 10.1523/jneurosci.0791-13.2013] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 04/08/2013] [Accepted: 04/11/2013] [Indexed: 11/21/2022] Open
Abstract
The mammalian neocortical progenitor cell niche is composed of a diverse repertoire of neuroepithelial cells, radial glia (RG), and intermediate neurogenic progenitors (INPs). Previously, live-cell imaging experiments have proved crucial in identifying these distinct progenitor populations, especially INPs, which amplify neural output by undergoing additional rounds of proliferation before differentiating into new neurons. INPs also provide feedback to the RG pool by serving as a source of Delta-like 1 (Dll1), a key ligand for activating Notch signaling in neighboring cells, a well-known mechanism for maintaining RG identity. While much is known about Dll1-Notch signaling at the molecular level, little is known about how this cell-cell contact dependent feedback is transmitted at the cellular level. To investigate how RG and INPs might interact to convey Notch signals, we used high-resolution live-cell multiphoton microscopy (MPM) to directly observe cellular interactions and dynamics, in conjunction with Notch-pathway specific reporters in the neocortical neural stem cell niche in organotypic brain slices from embryonic mice. We found that INPs and RG interact via dynamic and transient elongate processes, some apparently long-range (extending from the subventricular zone to the ventricular zone), and some short-range (filopodia-like). Gene expression profiling of RG and INPs revealed further progenitor cell diversification, including different subpopulations of Hes1+ and/or Hes5+ RG, and Dll1+ and/or Dll3+ INPs. Thus, the embryonic progenitor niche includes a network of dynamic cell-cell interactions, using different combinations of Notch signaling molecules to maintain and likely diversify progenitor pools.
Collapse
Affiliation(s)
- Branden R. Nelson
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington 98101, and
- Department of Neurological Surgery, University of Washington, Seattle, Washington 98101
| | - Rebecca D. Hodge
- Department of Neurological Surgery, University of Washington, Seattle, Washington 98101
| | - Francesco Bedogni
- Department of Neurological Surgery, University of Washington, Seattle, Washington 98101
| | - Robert F. Hevner
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington 98101, and
- Department of Neurological Surgery, University of Washington, Seattle, Washington 98101
| |
Collapse
|
46
|
Tuoc TC, Boretius S, Sansom SN, Pitulescu ME, Frahm J, Livesey FJ, Stoykova A. Chromatin regulation by BAF170 controls cerebral cortical size and thickness. Dev Cell 2013; 25:256-69. [PMID: 23643363 DOI: 10.1016/j.devcel.2013.04.005] [Citation(s) in RCA: 136] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 02/21/2013] [Accepted: 04/07/2013] [Indexed: 11/26/2022]
Abstract
Increased cortical size is essential to the enhanced intellectual capacity of primates during mammalian evolution. The mechanisms that control cortical size are largely unknown. Here, we show that mammalian BAF170, a subunit of the chromatin remodeling complex mSWI/SNF, is an intrinsic factor that controls cortical size. We find that conditional deletion of BAF170 promotes indirect neurogenesis by increasing the pool of intermediate progenitors (IPs) and results in an enlarged cortex, whereas cortex-specific BAF170 overexpression results in the opposite phenotype. Mechanistically, BAF170 competes with BAF155 subunit in the BAF complex, affecting euchromatin structure and thereby modulating the binding efficiency of the Pax6/REST-corepressor complex to Pax6 target genes that regulate the generation of IPs and late cortical progenitors. Our findings reveal a molecular mechanism mediated by the mSWI/SNF chromatin-remodeling complex that controls cortical architecture.
Collapse
Affiliation(s)
- Tran Cong Tuoc
- Research Group of Molecular Developmental Neurobiology, Department of Molecular Cell Biology, Max-Planck-Institute for Biophysical Chemistry, 37077 Göttingen, Germany
| | | | | | | | | | | | | |
Collapse
|
47
|
MicroRNA-92b regulates the development of intermediate cortical progenitors in embryonic mouse brain. Proc Natl Acad Sci U S A 2013; 110:7056-61. [PMID: 23569256 DOI: 10.1073/pnas.1219385110] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cerebral cortical neurons arise from radial glia (direct neurogenesis) or from intermediate progenitors (indirect neurogenesis); intriguingly, the sizes of intermediate progenitor populations and the cortices they generate correlate across species. The generation of intermediate progenitors is regulated by the transcription factor Tbr2, whose expression marks these cells. We investigated how this mechanism might be controlled. We found that acute blockade of mature microRNA biosynthesis in murine cortical progenitors caused a rapid cell autonomous increase in numbers of Tbr2-expressing cells. Acute microRNA-92b (miR-92b) gain of function caused rapid reductions in numbers of Tbr2-expressing cells and proliferating intermediate progenitors. Acute miR-92b loss of function had opposite effects. These findings indicate that miR-92b limits the production of intermediate cortical progenitors.
Collapse
|
48
|
Itoh Y, Moriyama Y, Hasegawa T, Endo TA, Toyoda T, Gotoh Y. Scratch regulates neuronal migration onset via an epithelial-mesenchymal transition-like mechanism. Nat Neurosci 2013; 16:416-25. [PMID: 23434913 DOI: 10.1038/nn.3336] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 01/23/2013] [Indexed: 12/15/2022]
Abstract
During neocortical development, the neuroepithelial or neural precursor cells that commit to neuronal fate need to delaminate and start migration toward the pial surface. However, the mechanism that couples neuronal fate commitment to detachment from the neuroepithelium remains largely unknown. Here we show that Scratch1 and Scratch2, members of the Snail superfamily of transcription factors, are expressed upon neuronal fate commitment under the control of proneural genes and promote apical process detachment and radial migration in the developing mouse neocortex. Scratch-induced delamination from the apical surface was mediated by transcriptional repression of the adhesion molecule E-cadherin. These findings suggest that Scratch proteins constitute a molecular link between neuronal fate commitment and the onset of neuronal migration. On the basis of their similarity to proteins involved in the epithelial-mesenchymal transition (EMT), we propose that Scratch proteins mediate the conversion of neuroepithelial cells to migrating neurons or intermediate neuronal progenitors through an EMT-related mechanism.
Collapse
Affiliation(s)
- Yasuhiro Itoh
- Institute of Molecular and Cellular Biosciences, University of Tokyo, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
49
|
Ohtaka-Maruyama C, Hirai S, Miwa A, Heng JIT, Shitara H, Ishii R, Taya C, Kawano H, Kasai M, Nakajima K, Okado H. RP58 regulates the multipolar-bipolar transition of newborn neurons in the developing cerebral cortex. Cell Rep 2013; 3:458-71. [PMID: 23395638 DOI: 10.1016/j.celrep.2013.01.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 11/16/2012] [Accepted: 01/14/2013] [Indexed: 01/03/2023] Open
Abstract
Accumulating evidence suggests that many brain diseases are associated with defects in neuronal migration, suggesting that this step of neurogenesis is critical for brain organization. However, the molecular mechanisms underlying neuronal migration remain largely unknown. Here, we identified the zinc-finger transcriptional repressor RP58 as a key regulator of neuronal migration via multipolar-to-bipolar transition. RP58(-/-) neurons exhibited severe defects in the formation of leading processes and never shifted to the locomotion mode. Cre-mediated deletion of RP58 using in utero electroporation in RP58(flox/flox) mice revealed that RP58 functions in cell-autonomous multipolar-to-bipolar transition, independent of cell-cycle exit. Finally, we found that RP58 represses Ngn2 transcription to regulate the Ngn2-Rnd2 pathway; Ngn2 knockdown rescued migration defects of the RP58(-/-) neurons. Our findings highlight the critical role of RP58 in multipolar-to-bipolar transition via suppression of the Ngn2-Rnd2 pathway in the developing cerebral cortex.
Collapse
Affiliation(s)
- Chiaki Ohtaka-Maruyama
- Department of Brain Development and Neural Regeneration, Neural Development Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Arai Y, Huttner WB, Calegari F. Neural Stem Cells. Regen Med 2013. [DOI: 10.1007/978-94-007-5690-8_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|