1
|
Moffat A, Schuurmans C. The Control of Cortical Folding: Multiple Mechanisms, Multiple Models. Neuroscientist 2024; 30:704-722. [PMID: 37621149 PMCID: PMC11558946 DOI: 10.1177/10738584231190839] [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] [Indexed: 08/26/2023]
Abstract
The cerebral cortex develops through a carefully conscripted series of cellular and molecular events that culminate in the production of highly specialized neuronal and glial cells. During development, cortical neurons and glia acquire a precise cellular arrangement and architecture to support higher-order cognitive functioning. Decades of study using rodent models, naturally gyrencephalic animal models, human pathology specimens, and, recently, human cerebral organoids, reveal that rodents recapitulate some but not all the cellular and molecular features of human cortices. Whereas rodent cortices are smooth-surfaced or lissencephalic, larger mammals, including humans and nonhuman primates, have highly folded/gyrencephalic cortices that accommodate an expansion in neuronal mass and increase in surface area. Several genes have evolved to drive cortical gyrification, arising from gene duplications or de novo origins, or by alterations to the structure/function of ancestral genes or their gene regulatory regions. Primary cortical folds arise in stereotypical locations, prefigured by a molecular "blueprint" that is set up by several signaling pathways (e.g., Notch, Fgf, Wnt, PI3K, Shh) and influenced by the extracellular matrix. Mutations that affect neural progenitor cell proliferation and/or neurogenesis, predominantly of upper-layer neurons, perturb cortical gyrification. Below we review the molecular drivers of cortical folding and their roles in disease.
Collapse
Affiliation(s)
- Alexandra Moffat
- Sunnybrook Research Institute, Biological Sciences Platform, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Carol Schuurmans
- Sunnybrook Research Institute, Biological Sciences Platform, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
2
|
Li M, Yuan Y, Hou Z, Hao S, Jin L, Wang B. Human brain organoid: trends, evolution, and remaining challenges. Neural Regen Res 2024; 19:2387-2399. [PMID: 38526275 PMCID: PMC11090441 DOI: 10.4103/1673-5374.390972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/26/2023] [Accepted: 10/28/2023] [Indexed: 03/26/2024] Open
Abstract
Advanced brain organoids provide promising platforms for deciphering the cellular and molecular processes of human neural development and diseases. Although various studies and reviews have described developments and advancements in brain organoids, few studies have comprehensively summarized and analyzed the global trends in this area of neuroscience. To identify and further facilitate the development of cerebral organoids, we utilized bibliometrics and visualization methods to analyze the global trends and evolution of brain organoids in the last 10 years. First, annual publications, countries/regions, organizations, journals, authors, co-citations, and keywords relating to brain organoids were identified. The hotspots in this field were also systematically identified. Subsequently, current applications for brain organoids in neuroscience, including human neural development, neural disorders, infectious diseases, regenerative medicine, drug discovery, and toxicity assessment studies, are comprehensively discussed. Towards that end, several considerations regarding the current challenges in brain organoid research and future strategies to advance neuroscience will be presented to further promote their application in neurological research.
Collapse
Affiliation(s)
- Minghui Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yuhan Yuan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Zongkun Hou
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Shilei Hao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Liang Jin
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Bochu Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| |
Collapse
|
3
|
Xu C, Alameri A, Leong W, Johnson E, Chen Z, Xu B, Leong KW. Multiscale engineering of brain organoids for disease modeling. Adv Drug Deliv Rev 2024; 210:115344. [PMID: 38810702 PMCID: PMC11265575 DOI: 10.1016/j.addr.2024.115344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/23/2024] [Accepted: 05/25/2024] [Indexed: 05/31/2024]
Abstract
Brain organoids hold great potential for modeling human brain development and pathogenesis. They recapitulate certain aspects of the transcriptional trajectory, cellular diversity, tissue architecture and functions of the developing brain. In this review, we explore the engineering strategies to control the molecular-, cellular- and tissue-level inputs to achieve high-fidelity brain organoids. We review the application of brain organoids in neural disorder modeling and emerging bioengineering methods to improve data collection and feature extraction at multiscale. The integration of multiscale engineering strategies and analytical methods has significant potential to advance insight into neurological disorders and accelerate drug development.
Collapse
Affiliation(s)
- Cong Xu
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Alia Alameri
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Wei Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Emily Johnson
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Zaozao Chen
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Bin Xu
- Department of Psychiatry, Columbia University, New York, NY 10032, USA.
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
4
|
Singh A, Del-Valle-Anton L, de Juan Romero C, Zhang Z, Ortuño EF, Mahesh A, Espinós A, Soler R, Cárdenas A, Fernández V, Lusby R, Tiwari VK, Borrell V. Gene regulatory landscape of cerebral cortex folding. SCIENCE ADVANCES 2024; 10:eadn1640. [PMID: 38838158 DOI: 10.1126/sciadv.adn1640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 05/02/2024] [Indexed: 06/07/2024]
Abstract
Folding of the cerebral cortex is a key aspect of mammalian brain development and evolution, and defects are linked to severe neurological disorders. Primary folding occurs in highly stereotyped patterns that are predefined in the cortical germinal zones by a transcriptomic protomap. The gene regulatory landscape governing the emergence of this folding protomap remains unknown. We characterized the spatiotemporal dynamics of gene expression and active epigenetic landscape (H3K27ac) across prospective folds and fissures in ferret. Our results show that the transcriptomic protomap begins to emerge at early embryonic stages, and it involves cell-fate signaling pathways. The H3K27ac landscape reveals developmental cell-fate restriction and engages known developmental regulators, including the transcription factor Cux2. Manipulating Cux2 expression in cortical progenitors changed their proliferation and the folding pattern in ferret, caused by selective transcriptional changes as revealed by single-cell RNA sequencing analyses. Our findings highlight the key relevance of epigenetic mechanisms in defining the patterns of cerebral cortex folding.
Collapse
Affiliation(s)
- Aditi Singh
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queens University Belfast, Belfast BT9 7BL, UK
| | - Lucia Del-Valle-Anton
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Sant Joan d'Alacant 03550, Spain
| | - Camino de Juan Romero
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Sant Joan d'Alacant 03550, Spain
| | - Ziyi Zhang
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queens University Belfast, Belfast BT9 7BL, UK
| | - Eduardo Fernández Ortuño
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Sant Joan d'Alacant 03550, Spain
| | - Arun Mahesh
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queens University Belfast, Belfast BT9 7BL, UK
- Institute for Molecular Medicine, University of Southern Denmark, Odense M, Denmark
| | - Alexandre Espinós
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Sant Joan d'Alacant 03550, Spain
| | - Rafael Soler
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Sant Joan d'Alacant 03550, Spain
| | - Adrián Cárdenas
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Sant Joan d'Alacant 03550, Spain
| | - Virginia Fernández
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Sant Joan d'Alacant 03550, Spain
| | - Ryan Lusby
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queens University Belfast, Belfast BT9 7BL, UK
| | - Vijay K Tiwari
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry, and Biomedical Science, Queens University Belfast, Belfast BT9 7BL, UK
- Institute for Molecular Medicine, University of Southern Denmark, Odense M, Denmark
- Danish Institute for Advanced Study (DIAS), Odense M, Denmark
- Department of Clinical Genetics, Odense University Hospital, Odense C, Denmark
| | - Víctor Borrell
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Sant Joan d'Alacant 03550, Spain
| |
Collapse
|
5
|
LaMonica Ostrem BE, Domínguez-Iturza N, Stogsdill JA, Faits T, Kim K, Levin JZ, Arlotta P. Fetal brain response to maternal inflammation requires microglia. Development 2024; 151:dev202252. [PMID: 38775708 PMCID: PMC11190434 DOI: 10.1242/dev.202252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 04/09/2024] [Indexed: 06/23/2024]
Abstract
In utero infection and maternal inflammation can adversely impact fetal brain development. Maternal systemic illness, even in the absence of direct fetal brain infection, is associated with an increased risk of neuropsychiatric disorders in affected offspring. The cell types mediating the fetal brain response to maternal inflammation are largely unknown, hindering the development of novel treatment strategies. Here, we show that microglia, the resident phagocytes of the brain, highly express receptors for relevant pathogens and cytokines throughout embryonic development. Using a rodent maternal immune activation (MIA) model in which polyinosinic:polycytidylic acid is injected into pregnant mice, we demonstrate long-lasting transcriptional changes in fetal microglia that persist into postnatal life. We find that MIA induces widespread gene expression changes in neuronal and non-neuronal cells; importantly, these responses are abolished by selective genetic deletion of microglia, indicating that microglia are required for the transcriptional response of other cortical cell types to MIA. These findings demonstrate that microglia play a crucial durable role in the fetal response to maternal inflammation, and should be explored as potential therapeutic cell targets.
Collapse
Affiliation(s)
- Bridget Elaine LaMonica Ostrem
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Nuria Domínguez-Iturza
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jeffrey A. Stogsdill
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Tyler Faits
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kwanho Kim
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Joshua Z. Levin
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Paola Arlotta
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
6
|
Prajapati K, Yan C, Yang Q, Arbitman S, Fitzgerald DP, Sharee S, Shaik J, Bosiacki J, Myers K, Paucarmayta A, Johnson DM, O’Neill T, Kundu S, Cusumano Z, Langermann S, Langenau DM, Patel S, Flies DB. The FLRT3-UNC5B checkpoint pathway inhibits T cell-based cancer immunotherapies. SCIENCE ADVANCES 2024; 10:eadj4698. [PMID: 38427724 PMCID: PMC10906930 DOI: 10.1126/sciadv.adj4698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 01/25/2024] [Indexed: 03/03/2024]
Abstract
Cancers exploit coinhibitory receptors on T cells to escape tumor immunity, and targeting such mechanisms has shown remarkable clinical benefit, but in a limited subset of patients. We hypothesized that cancer cells mimic noncanonical mechanisms of early development such as axon guidance pathways to evade T cell immunity. Using gain-of-function genetic screens, we profiled axon guidance proteins on human T cells and their cognate ligands and identified fibronectin leucine-rich transmembrane protein 3 (FLRT3) as a ligand that inhibits T cell activity. We demonstrated that FLRT3 inhibits T cells through UNC5B, an axon guidance receptor that is up-regulated on activated human T cells. FLRT3 expressed in human cancers favored tumor growth and inhibited CAR-T and BiTE + T cell killing and infiltration in humanized cancer models. An FLRT3 monoclonal antibody that blocked FLRT3-UNC5B interactions reversed these effects in an immune-dependent manner. This study supports the concept that axon guidance proteins mimic T cell checkpoints and can be targeted for cancer immunotherapy.
Collapse
Affiliation(s)
| | - Chuan Yan
- Molecular Pathology and Cancer Center, Massachusetts General Hospital Research Institute, Charlestown, MA 02129, USA
- Harvard Stem Cell Institute, Cambridge, MA 02139, USA
| | - Qiqi Yang
- Molecular Pathology and Cancer Center, Massachusetts General Hospital Research Institute, Charlestown, MA 02129, USA
- Harvard Stem Cell Institute, Cambridge, MA 02139, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | - David M. Langenau
- Molecular Pathology and Cancer Center, Massachusetts General Hospital Research Institute, Charlestown, MA 02129, USA
- Harvard Stem Cell Institute, Cambridge, MA 02139, USA
| | | | | |
Collapse
|
7
|
Barresi M, Hickmott RA, Bosakhar A, Quezada S, Quigley A, Kawasaki H, Walker D, Tolcos M. Toward a better understanding of how a gyrified brain develops. Cereb Cortex 2024; 34:bhae055. [PMID: 38425213 DOI: 10.1093/cercor/bhae055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 01/26/2024] [Accepted: 01/28/2024] [Indexed: 03/02/2024] Open
Abstract
The size and shape of the cerebral cortex have changed dramatically across evolution. For some species, the cortex remains smooth (lissencephalic) throughout their lifetime, while for other species, including humans and other primates, the cortex increases substantially in size and becomes folded (gyrencephalic). A folded cortex boasts substantially increased surface area, cortical thickness, and neuronal density, and it is therefore associated with higher-order cognitive abilities. The mechanisms that drive gyrification in some species, while others remain lissencephalic despite many shared neurodevelopmental features, have been a topic of investigation for many decades, giving rise to multiple perspectives of how the gyrified cerebral cortex acquires its unique shape. Recently, a structurally unique germinal layer, known as the outer subventricular zone, and the specialized cell type that populates it, called basal radial glial cells, were identified, and these have been shown to be indispensable for cortical expansion and folding. Transcriptional analyses and gene manipulation models have provided an invaluable insight into many of the key cellular and genetic drivers of gyrification. However, the degree to which certain biomechanical, genetic, and cellular processes drive gyrification remains under investigation. This review considers the key aspects of cerebral expansion and folding that have been identified to date and how theories of gyrification have evolved to incorporate this new knowledge.
Collapse
Affiliation(s)
- Mikaela Barresi
- School of Health and Biomedical Sciences, RMIT University, Plenty Road, Bundoora, VIC 3083, Australia
- ACMD, St Vincent's Hospital Melbourne, Regent Street, Fitzroy, VIC 3065, Australia
| | - Ryan Alexander Hickmott
- School of Health and Biomedical Sciences, RMIT University, Plenty Road, Bundoora, VIC 3083, Australia
- ACMD, St Vincent's Hospital Melbourne, Regent Street, Fitzroy, VIC 3065, Australia
| | - Abdulhameed Bosakhar
- School of Health and Biomedical Sciences, RMIT University, Plenty Road, Bundoora, VIC 3083, Australia
| | - Sebastian Quezada
- School of Health and Biomedical Sciences, RMIT University, Plenty Road, Bundoora, VIC 3083, Australia
| | - Anita Quigley
- School of Health and Biomedical Sciences, RMIT University, Plenty Road, Bundoora, VIC 3083, Australia
- ACMD, St Vincent's Hospital Melbourne, Regent Street, Fitzroy, VIC 3065, Australia
- School of Engineering, RMIT University, La Trobe Street, Melbourne, VIC 3000, Australia
- Department of Medicine, University of Melbourne, St Vincent's Hospital, Regent Street, Fitzroy, VIC 3065, Australia
| | - Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Takara-machi 13-1, Kanazawa, Ishikawa 920-8640, Japan
| | - David Walker
- School of Health and Biomedical Sciences, RMIT University, Plenty Road, Bundoora, VIC 3083, Australia
| | - Mary Tolcos
- School of Health and Biomedical Sciences, RMIT University, Plenty Road, Bundoora, VIC 3083, Australia
| |
Collapse
|
8
|
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
|
9
|
Akula SK, Exposito-Alonso D, Walsh CA. Shaping the brain: The emergence of cortical structure and folding. Dev Cell 2023; 58:2836-2849. [PMID: 38113850 PMCID: PMC10793202 DOI: 10.1016/j.devcel.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 04/08/2023] [Accepted: 11/10/2023] [Indexed: 12/21/2023]
Abstract
The cerebral cortex-the brain's covering and largest region-has increased in size and complexity in humans and supports higher cognitive functions such as language and abstract thinking. There is a growing understanding of the human cerebral cortex, including the diversity and number of cell types that it contains, as well as of the developmental mechanisms that shape cortical structure and organization. In this review, we discuss recent progress in our understanding of molecular and cellular processes, as well as mechanical forces, that regulate the folding of the cerebral cortex. Advances in human genetics, coupled with experimental modeling in gyrencephalic species, have provided insights into the central role of cortical progenitors in the gyrification and evolutionary expansion of the cerebral cortex. These studies are essential for understanding the emergence of structural and functional organization during cortical development and the pathogenesis of neurodevelopmental disorders associated with cortical malformations.
Collapse
Affiliation(s)
- Shyam K Akula
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - David Exposito-Alonso
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Christopher A Walsh
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA; Departments of Pediatrics and Neurology, Harvard Medical School, Boston, MA, USA; Allen Discovery Center for Human Brain Evolution, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Chevy Chase, Maryland, USA.
| |
Collapse
|
10
|
Zhou Z, Pan Y, Zhou S, Wang S, Zhang D, Cao Y, Jiang X, Li J, Zhu L, Zhao L, Gu S, Lin G, Dong Z, Sun HX. Single-cell analysis reveals specific neuronal transition during mouse corticogenesis. Front Cell Dev Biol 2023; 11:1209320. [PMID: 38020907 PMCID: PMC10657809 DOI: 10.3389/fcell.2023.1209320] [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: 04/20/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Background: Currently, the mechanism(s) underlying corticogenesis is still under characterization. Methods: We curated the most comprehensive single-cell RNA-seq (scRNA-seq) datasets from mouse and human fetal cortexes for data analysis and confirmed the findings with co-immunostaining experiments. Results: By analyzing the developmental trajectories with scRNA-seq datasets in mice, we identified a specific developmental sub-path contributed by a cell-population expressing both deep- and upper-layer neurons (DLNs and ULNs) specific markers, which occurred on E13.5 but was absent in adults. In this cell-population, the percentages of cells expressing DLN and ULN markers decreased and increased, respectively, during the development suggesting direct neuronal transition (namely D-T-U). Whilst genes significantly highly/uniquely expressed in D-T-U cell population were significantly enriched in PTN/MDK signaling pathways related to cell migration. Both findings were further confirmed by co-immunostaining with DLNs, ULNs and D-T-U specific markers across different timepoints. Furthermore, six genes (co-expressed with D-T-U specific markers in mice) showing a potential opposite temporal expression between human and mouse during fetal cortical development were associated with neuronal migration and cognitive functions. In adult prefrontal cortexes (PFC), D-T-U specific genes were expressed in neurons from different layers between humans and mice. Conclusion: Our study characterizes a specific cell population D-T-U showing direct DLNs to ULNs neuronal transition and migration during fetal cortical development in mice. It is potentially associated with the difference of cortical development in humans and mice.
Collapse
Affiliation(s)
- Ziheng Zhou
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Yueyang Pan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Si Zhou
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Shuguang Wang
- Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Dengwei Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Ye Cao
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Xiaosen Jiang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jie Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Linnan Zhu
- Biomedical Pioneering Innovation Center (BIOPIC), Academy for Advanced Interdisciplinary Studies, School of Life Sciences, Peking University, Beijing, China
| | - Lijian Zhao
- Medical Technology College, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Shen Gu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Ge Lin
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Zirui Dong
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
| | - Hai-Xi Sun
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
11
|
Prigge CL, Dembla M, Sharma A, El-Quessny M, Kozlowski C, Paisley CE, Miltner AM, Johnson TM, Della Santina L, Feller MB, Kay JN. Rejection of inappropriate synaptic partners in mouse retina mediated by transcellular FLRT2-UNC5 signaling. Dev Cell 2023; 58:2080-2096.e7. [PMID: 37557174 PMCID: PMC10615732 DOI: 10.1016/j.devcel.2023.07.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 05/26/2023] [Accepted: 07/18/2023] [Indexed: 08/11/2023]
Abstract
During nervous system development, neurons choose synaptic partners with remarkable specificity; however, the cell-cell recognition mechanisms governing rejection of inappropriate partners remain enigmatic. Here, we show that mouse retinal neurons avoid inappropriate partners by using the FLRT2-uncoordinated-5 (UNC5) receptor-ligand system. Within the inner plexiform layer (IPL), FLRT2 is expressed by direction-selective (DS) circuit neurons, whereas UNC5C/D are expressed by non-DS neurons projecting to adjacent IPL sublayers. In vivo gain- and loss-of-function experiments demonstrate that FLRT2-UNC5 binding eliminates growing DS dendrites that have strayed from the DS circuit IPL sublayers. Abrogation of FLRT2-UNC5 binding allows mistargeted arbors to persist, elaborate, and acquire synapses from inappropriate partners. Conversely, UNC5C misexpression within DS circuit sublayers inhibits dendrite growth and drives arbors into adjacent sublayers. Mechanistically, UNC5s promote dendrite elimination by interfering with FLRT2-mediated adhesion. Based on their broad expression, FLRT-UNC5 recognition is poised to exert widespread effects upon synaptic partner choices across the nervous system.
Collapse
Affiliation(s)
- Cameron L Prigge
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Mayur Dembla
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Arsha Sharma
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Malak El-Quessny
- Helen Wills Neuroscience Institute and Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Christopher Kozlowski
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Caitlin E Paisley
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Adam M Miltner
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Tyler M Johnson
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Luca Della Santina
- Department of Vision Sciences, University of Houston College of Optometry, Houston, TX 77204, USA
| | - Marla B Feller
- Helen Wills Neuroscience Institute and Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jeremy N Kay
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA.
| |
Collapse
|
12
|
Matrongolo MJ, Ho-Nguyen KT, Jain M, Ang PS, Reddy A, Schaper S, Tischfield MA. Loss of Twist1 and balanced retinoic acid signaling from the meninges causes cortical folding in mice. Development 2023; 150:dev201381. [PMID: 37590085 PMCID: PMC11296311 DOI: 10.1242/dev.201381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 08/08/2023] [Indexed: 08/18/2023]
Abstract
Secondary lissencephaly evolved in mice due to effects on neurogenesis and the tangential distribution of neurons. Signaling pathways that help maintain lissencephaly are still poorly understood. We show that inactivating Twist1 in the primitive meninges causes cortical folding in mice. Cell proliferation in the meninges is reduced, causing loss of arachnoid fibroblasts that express Raldh2, an enzyme required for retinoic acid synthesis. Regionalized loss of Raldh2 in the dorsolateral meninges is first detected when folding begins. The ventricular zone expands and the forebrain lengthens at this time due to expansion of apical radial glia. As the cortex expands, regionalized differences in the levels of neurogenesis are coupled with changes to the tangential distribution of neurons. Consequentially, cortical growth at and adjacent to the midline accelerates with respect to more dorsolateral regions, resulting in cortical buckling and folding. Maternal retinoic acid supplementation suppresses cortical folding by normalizing forebrain length, neurogenesis and the tangential distribution of neurons. These results suggest that Twist1 and balanced retinoic acid signaling from the meninges are required to maintain normal levels of neurogenesis and lissencephaly in mice.
Collapse
Affiliation(s)
- Matt J. Matrongolo
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Khue-Tu Ho-Nguyen
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Manav Jain
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Phillip S. Ang
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
- University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
| | - Akash Reddy
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Samantha Schaper
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Max A. Tischfield
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| |
Collapse
|
13
|
Toudji I, Toumi A, Chamberland É, Rossignol E. Interneuron odyssey: molecular mechanisms of tangential migration. Front Neural Circuits 2023; 17:1256455. [PMID: 37779671 PMCID: PMC10538647 DOI: 10.3389/fncir.2023.1256455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/21/2023] [Indexed: 10/03/2023] Open
Abstract
Cortical GABAergic interneurons are critical components of neural networks. They provide local and long-range inhibition and help coordinate network activities involved in various brain functions, including signal processing, learning, memory and adaptative responses. Disruption of cortical GABAergic interneuron migration thus induces profound deficits in neural network organization and function, and results in a variety of neurodevelopmental and neuropsychiatric disorders including epilepsy, intellectual disability, autism spectrum disorders and schizophrenia. It is thus of paramount importance to elucidate the specific mechanisms that govern the migration of interneurons to clarify some of the underlying disease mechanisms. GABAergic interneurons destined to populate the cortex arise from multipotent ventral progenitor cells located in the ganglionic eminences and pre-optic area. Post-mitotic interneurons exit their place of origin in the ventral forebrain and migrate dorsally using defined migratory streams to reach the cortical plate, which they enter through radial migration before dispersing to settle in their final laminar allocation. While migrating, cortical interneurons constantly change their morphology through the dynamic remodeling of actomyosin and microtubule cytoskeleton as they detect and integrate extracellular guidance cues generated by neuronal and non-neuronal sources distributed along their migratory routes. These processes ensure proper distribution of GABAergic interneurons across cortical areas and lamina, supporting the development of adequate network connectivity and brain function. This short review summarizes current knowledge on the cellular and molecular mechanisms controlling cortical GABAergic interneuron migration, with a focus on tangential migration, and addresses potential avenues for cell-based interneuron progenitor transplants in the treatment of neurodevelopmental disorders and epilepsy.
Collapse
Affiliation(s)
- Ikram Toudji
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Montréal, QC, Canada
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Asmaa Toumi
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Montréal, QC, Canada
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC, Canada
| | - Émile Chamberland
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Montréal, QC, Canada
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Elsa Rossignol
- Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Montréal, QC, Canada
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
- Department of Pediatrics, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
14
|
Ritter C, Eigen L, Deiringer N, Laubscher L, Brecht M. Coevolution of rostrum and brain in pig species. J Comp Neurol 2023; 531:775-789. [PMID: 36843325 DOI: 10.1002/cne.25461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 01/17/2023] [Accepted: 01/23/2023] [Indexed: 02/28/2023]
Abstract
Domestic pigs have a prominent cortical gyrus (the rostrum gyrus) isomorphic to the contralateral hemirostrum. It is unclear, however, if the size and shape of the rostrum gyrus are of evolutionary/functional relevance. Here, we address this question by assessing the relationship of rostrum and rostrum gyrus across eight pig species. To this end, we quantified rostrum morphology in fresh and alcohol-preserved pig specimens by surface scans, microfocus computed tomography scans, and photography. We establish that the size and shape of the rostrum gyrus can be precisely inferred from endocasts. We then took advantage of the accessibility of pig skulls and endocasts to assess features of the rostrum gyrus across species. Our investigation led to the following results: (i) The rostra of pig species show basic similarities. (ii) A cortical rostrum gyrus is apparent in all pigs. (iii) The size of the rostrum gyrus differs across species and outgroups of the evolutionary dominant suinae (i.e., peccaries and the babirusa) have a noticeably smaller rostrum gyrus. (iv) Warthogs have a derived rostrum morphology with an extra fold and a very wide rostrum; the warthog rostrum gyrus recapitulates these rostrum features. (v) Domestic pigs have relatively smaller rostrum gyrus than wild boars. We also provide indications for a conserved cytoarchitectonic patterning of the rostrum gyrus. We conclude that the rostrum gyrus is a neural module that was putatively present in the common ancestor of pigs and that this neural module is conserved across pig species. Natural selection maintains the rostrum gyrus' size and its exact isomorphism to the rostrum.
Collapse
Affiliation(s)
- Cindy Ritter
- Bernstein Center for Computational Neuroscience Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lennart Eigen
- Bernstein Center for Computational Neuroscience Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nora Deiringer
- Bernstein Center for Computational Neuroscience Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Liesel Laubscher
- Wildlife Pharmaceuticals Wildlife Research Facility, Wildlife Pharmaceuticals (Pty) Ltd, White River, South Africa
| | - Michael Brecht
- Bernstein Center for Computational Neuroscience Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
15
|
Fang Y, Ma K, Huang YM, Dang Y, Liu Z, Xu Y, Zheng XL, Yang X, Huo Y, Dai X. Fibronectin leucine-rich transmembrane protein 2 drives monocyte differentiation into macrophages via the UNC5B-Akt/mTOR axis. Front Immunol 2023; 14:1162004. [PMID: 37090697 PMCID: PMC10117657 DOI: 10.3389/fimmu.2023.1162004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/27/2023] [Indexed: 04/09/2023] Open
Abstract
Upon migrating into the tissues, hematopoietic stem cell (HSC)-derived monocytes differentiate into macrophages, playing a crucial role in determining innate immune responses towards external pathogens and internal stimuli. However, the regulatory mechanisms underlying monocyte-to-macrophage differentiation remain largely unexplored. Here we divulge a previously uncharacterized but essential role for an axon guidance molecule, fibronectin leucine-rich transmembrane protein 2 (FLRT2), in monocyte-to-macrophage maturation. FLRT2 is almost undetectable in human monocytic cell lines, human peripheral blood mononuclear cells (PBMCs), and mouse primary monocytes but significantly increases in fully differentiated macrophages. Myeloid-specific deletion of FLRT2 (Flrt2ΔMyel) contributes to decreased peritoneal monocyte-to-macrophage generation in mice in vivo, accompanied by impaired macrophage functions. Gain- and loss-of-function studies support the promoting effect of FLRT2 on THP-1 cell and human PBMC differentiation into macrophages. Mechanistically, FLRT2 directly interacts with Unc-5 netrin receptor B (UNC5B) via its extracellular domain (ECD) and activates Akt/mTOR signaling. In vivo administration of mTOR agonist MYH1485 reverses the impaired phenotypes observed in Flrt2ΔMyel mice. Together, these results identify FLRT2 as a novel pivotal endogenous regulator of monocyte differentiation into macrophages. Targeting the FLRT2/UNC5B-Akt/mTOR axis may provide potential therapeutic strategies directly relevant to human diseases associated with aberrant monocyte/macrophage differentiation.
Collapse
Affiliation(s)
- Yaxiong Fang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Kongyang Ma
- Centre for Infection and Immunity Studies (CIIS), School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yi-Min Huang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yuanye Dang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhaoyu Liu
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yiming Xu
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xi-Long Zheng
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Xiangdong Yang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yongliang Huo
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Experimental Animal Center, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
- *Correspondence: Xiaoyan Dai, ; Yongliang Huo,
| | - Xiaoyan Dai
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
- *Correspondence: Xiaoyan Dai, ; Yongliang Huo,
| |
Collapse
|
16
|
Qi J, Mo F, An NA, Mi T, Wang J, Qi J, Li X, Zhang B, Xia L, Lu Y, Sun G, Wang X, Li C, Hu B. A Human-Specific De Novo Gene Promotes Cortical Expansion and Folding. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204140. [PMID: 36638273 PMCID: PMC9982566 DOI: 10.1002/advs.202204140] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 12/20/2022] [Indexed: 06/17/2023]
Abstract
Newly originated de novo genes have been linked to the formation and function of the human brain. However, how a specific gene originates from ancestral noncoding DNAs and becomes involved in the preexisting network for functional outcomes remains elusive. Here, a human-specific de novo gene, SP0535, is identified that is preferentially expressed in the ventricular zone of the human fetal brain and plays an important role in cortical development and function. In human embryonic stem cell-derived cortical organoids, knockout of SP0535 compromises their growth and neurogenesis. In SP0535 transgenic (TG) mice, expression of SP0535 induces fetal cortex expansion and sulci and gyri-like structure formation. The progenitors and neurons in the SP0535 TG mouse cortex tend to proliferate and differentiate in ways that are unique to humans. SP0535 TG adult mice also exhibit improved cognitive ability and working memory. Mechanistically, SP0535 interacts with the membrane protein Na+ /K+ ATPase subunit alpha-1 (ATP1A1) and releases Src from the ATP1A1-Src complex, allowing increased level of Src phosphorylation that promotes cell proliferation. Thus, SP0535 is the first proven human-specific de novo gene that promotes cortical expansion and folding, and can function through incorporating into an existing conserved molecular network.
Collapse
Affiliation(s)
- Jianhuan Qi
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Savaid Medical SchoolUniversity of Chinese Academy of SciencesBeijing100049China
| | - Fan Mo
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Savaid Medical SchoolUniversity of Chinese Academy of SciencesBeijing100049China
| | - Ni A. An
- Laboratory of Bioinformatics and Genomic MedicineInstitute of Molecular MedicineCollege of Future TechnologyPeking UniversityBeijing100871China
| | - Tingwei Mi
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
| | - Jiaxin Wang
- Laboratory of Bioinformatics and Genomic MedicineInstitute of Molecular MedicineCollege of Future TechnologyPeking UniversityBeijing100871China
| | - Jun‐Tian Qi
- Laboratory of Bioinformatics and Genomic MedicineInstitute of Molecular MedicineCollege of Future TechnologyPeking UniversityBeijing100871China
| | - Xiangshang Li
- Laboratory of Bioinformatics and Genomic MedicineInstitute of Molecular MedicineCollege of Future TechnologyPeking UniversityBeijing100871China
| | - Boya Zhang
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
| | - Longkuo Xia
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Savaid Medical SchoolUniversity of Chinese Academy of SciencesBeijing100049China
| | - Yingfei Lu
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Savaid Medical SchoolUniversity of Chinese Academy of SciencesBeijing100049China
| | - Gaoying Sun
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Savaid Medical SchoolUniversity of Chinese Academy of SciencesBeijing100049China
| | - Xinyue Wang
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Savaid Medical SchoolUniversity of Chinese Academy of SciencesBeijing100049China
| | - Chuan‐Yun Li
- Laboratory of Bioinformatics and Genomic MedicineInstitute of Molecular MedicineCollege of Future TechnologyPeking UniversityBeijing100871China
| | - Baoyang Hu
- State Key Laboratory of Stem Cell and Reproductive BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- Savaid Medical SchoolUniversity of Chinese Academy of SciencesBeijing100049China
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijing100101China
- Beijing Institute for Stem Cell and Regenerative MedicineBeijing100101China
| |
Collapse
|
17
|
Vaid S, Heikinheimo O, Namba T. Embryonic mouse medial neocortex as a model system for studying the radial glial scaffold in fetal human neocortex. J Neural Transm (Vienna) 2023; 130:185-194. [PMID: 36450874 PMCID: PMC10033555 DOI: 10.1007/s00702-022-02570-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/18/2022] [Indexed: 12/03/2022]
Abstract
Neocortex is the evolutionarily newest region in the brain, and is a structure with diversified size and morphology among mammalian species. Humans have the biggest neocortex compared to the body size, and their neocortex has many foldings, that is, gyri and sulci. Despite the recent methodological advances in in vitro models such as cerebral organoids, mice have been continuously used as a model system for studying human neocortical development because of the accessibility and practicality of in vivo gene manipulation. The commonly studied neocortical region, the lateral neocortex, generally recapitulates the developmental process of the human neocortex, however, there are several important factors missing in the lateral neocortex. First, basal (outer) radial glia (bRG), which are the main cell type providing the radial scaffold to the migrating neurons in the fetal human neocortex, are very few in the mouse lateral neocortex, thus the radial glial scaffold is different from the fetal human neocortex. Second, as a consequence of the difference in the radial glial scaffold, migrating neurons might exhibit different migratory behavior and thus distribution. To overcome those problems, we propose the mouse medial neocortex, where we have earlier revealed an abundance of bRG similar to the fetal human neocortex, as an alternative model system. We found that similar to the fetal human neocortex, the radial glial scaffold, neuronal migration and neuronal distribution are tangentially scattered in the mouse medial neocortex. Taken together, the embryonic mouse medial neocortex could be a suitable and accessible in vivo model system to study human neocortical development and its pathogenesis.
Collapse
Affiliation(s)
- Samir Vaid
- Department of Basic Neurosciences, University of Geneva, 1211, Geneva, Switzerland
| | - Oskari Heikinheimo
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, P.O. 140, 00029, Helsinki, Finland
| | - Takashi Namba
- Neuroscience Center, HiLIFE - Helsinki Institute of Life Science, University of Helsinki, P.O. 63, 00014, Helsinki, Finland.
| |
Collapse
|
18
|
Damianidou E, Mouratidou L, Kyrousi C. Research models of neurodevelopmental disorders: The right model in the right place. Front Neurosci 2022; 16:1031075. [PMID: 36340790 PMCID: PMC9630472 DOI: 10.3389/fnins.2022.1031075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/07/2022] [Indexed: 11/25/2022] Open
Abstract
Neurodevelopmental disorders (NDDs) are a heterogeneous group of impairments that affect the development of the central nervous system leading to abnormal brain function. NDDs affect a great percentage of the population worldwide, imposing a high societal and economic burden and thus, interest in this field has widely grown in recent years. Nevertheless, the complexity of human brain development and function as well as the limitations regarding human tissue usage make their modeling challenging. Animal models play a central role in the investigation of the implicated molecular and cellular mechanisms, however many of them display key differences regarding human phenotype and in many cases, they partially or completely fail to recapitulate them. Although in vitro two-dimensional (2D) human-specific models have been highly used to address some of these limitations, they lack crucial features such as complexity and heterogeneity. In this review, we will discuss the advantages, limitations and future applications of in vivo and in vitro models that are used today to model NDDs. Additionally, we will describe the recent development of 3-dimensional brain (3D) organoids which offer a promising approach as human-specific in vitro models to decipher these complex disorders.
Collapse
Affiliation(s)
- Eleni Damianidou
- University Mental Health, Neurosciences and Precision Medicine Research Institute “Costas Stefanis”, Athens, Greece
| | - Lidia Mouratidou
- University Mental Health, Neurosciences and Precision Medicine Research Institute “Costas Stefanis”, Athens, Greece
- First Department of Psychiatry, Medical School, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Kyrousi
- University Mental Health, Neurosciences and Precision Medicine Research Institute “Costas Stefanis”, Athens, Greece
- First Department of Psychiatry, Medical School, Eginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
- *Correspondence: Christina Kyrousi,
| |
Collapse
|
19
|
Akkermans O, Delloye-Bourgeois C, Peregrina C, Carrasquero-Ordaz M, Kokolaki M, Berbeira-Santana M, Chavent M, Reynaud F, Raj R, Agirre J, Aksu M, White ES, Lowe E, Ben Amar D, Zaballa S, Huo J, Pakos I, McCubbin PTN, Comoletti D, Owens RJ, Robinson CV, Castellani V, Del Toro D, Seiradake E. GPC3-Unc5 receptor complex structure and role in cell migration. Cell 2022; 185:3931-3949.e26. [PMID: 36240740 PMCID: PMC9596381 DOI: 10.1016/j.cell.2022.09.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 07/22/2022] [Accepted: 09/15/2022] [Indexed: 11/09/2022]
Abstract
Neural migration is a critical step during brain development that requires the interactions of cell-surface guidance receptors. Cancer cells often hijack these mechanisms to disseminate. Here, we reveal crystal structures of Uncoordinated-5 receptor D (Unc5D) in complex with morphogen receptor glypican-3 (GPC3), forming an octameric glycoprotein complex. In the complex, four Unc5D molecules pack into an antiparallel bundle, flanked by four GPC3 molecules. Central glycan-glycan interactions are formed by N-linked glycans emanating from GPC3 (N241 in human) and C-mannosylated tryptophans of the Unc5D thrombospondin-like domains. MD simulations, mass spectrometry and structure-based mutants validate the crystallographic data. Anti-GPC3 nanobodies enhance or weaken Unc5-GPC3 binding and, together with mutant proteins, show that Unc5/GPC3 guide migrating pyramidal neurons in the mouse cortex, and cancer cells in an embryonic xenograft neuroblastoma model. The results demonstrate a conserved structural mechanism of cell guidance, where finely balanced Unc5-GPC3 interactions regulate cell migration.
Collapse
Affiliation(s)
- Onno Akkermans
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Céline Delloye-Bourgeois
- MeLis, University of Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5284, INSERM U1314, Institut NeuroMyoGène, 8 avenue Rockefeller 69008 Lyon, Lyon, France
| | - Claudia Peregrina
- Department of Biological Sciences, Institute of Neurosciences, IDIBAPS, CIBERNED, University of Barcelona, Barcelona, Spain
| | - Maria Carrasquero-Ordaz
- Department of Biochemistry, University of Oxford, Oxford, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Maria Kokolaki
- Department of Biochemistry, University of Oxford, Oxford, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Miguel Berbeira-Santana
- Department of Biochemistry, University of Oxford, Oxford, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Matthieu Chavent
- Institut de Pharmacologie et Biologie Structurale, Université de Toulouse, Toulouse, France
| | - Florie Reynaud
- MeLis, University of Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5284, INSERM U1314, Institut NeuroMyoGène, 8 avenue Rockefeller 69008 Lyon, Lyon, France
| | - Ritu Raj
- Department of Chemistry, University of Oxford, Oxford, UK
| | - Jon Agirre
- York Structural Biology Laboratory, Department of Chemistry, University of York, York, UK
| | - Metin Aksu
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Eleanor S White
- Department of Biochemistry, University of Oxford, Oxford, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Edward Lowe
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Dounia Ben Amar
- MeLis, University of Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5284, INSERM U1314, Institut NeuroMyoGène, 8 avenue Rockefeller 69008 Lyon, Lyon, France
| | - Sofia Zaballa
- Department of Biological Sciences, Institute of Neurosciences, IDIBAPS, CIBERNED, University of Barcelona, Barcelona, Spain
| | - Jiandong Huo
- Structural Biology, The Rosalind Franklin Institute, Harwell Science Campus, Didcot, UK; Division of Structural Biology, University of Oxford, Oxford, UK
| | - Irene Pakos
- Child Health Institute of New Jersey, New Brunswick, NJ 08901, USA
| | - Patrick T N McCubbin
- Department of Biochemistry, University of Oxford, Oxford, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Davide Comoletti
- Child Health Institute of New Jersey, New Brunswick, NJ 08901, USA; School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Raymond J Owens
- Structural Biology, The Rosalind Franklin Institute, Harwell Science Campus, Didcot, UK; Division of Structural Biology, University of Oxford, Oxford, UK
| | - Carol V Robinson
- Department of Chemistry, University of Oxford, Oxford, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Valérie Castellani
- MeLis, University of Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5284, INSERM U1314, Institut NeuroMyoGène, 8 avenue Rockefeller 69008 Lyon, Lyon, France.
| | - Daniel Del Toro
- Department of Biological Sciences, Institute of Neurosciences, IDIBAPS, CIBERNED, University of Barcelona, Barcelona, Spain.
| | - Elena Seiradake
- Department of Biochemistry, University of Oxford, Oxford, UK; Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK.
| |
Collapse
|
20
|
Massimo M, Long KR. Orchestrating human neocortex development across the scales; from micro to macro. Semin Cell Dev Biol 2022; 130:24-36. [PMID: 34583893 DOI: 10.1016/j.semcdb.2021.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 08/27/2021] [Accepted: 09/10/2021] [Indexed: 10/20/2022]
Abstract
How our brains have developed to perform the many complex functions that make us human has long remained a question of great interest. Over the last few decades, many scientists from a wide range of fields have tried to answer this question by aiming to uncover the mechanisms that regulate the development of the human neocortex. They have approached this on different scales, focusing microscopically on individual cells all the way up to macroscopically imaging entire brains within living patients. In this review we will summarise these key findings and how they fit together.
Collapse
Affiliation(s)
- Marco Massimo
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, United Kingdom; MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, United Kingdom
| | - Katherine R Long
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE1 1UL, United Kingdom; MRC Centre for Neurodevelopmental Disorders, King's College London, London SE1 1UL, United Kingdom.
| |
Collapse
|
21
|
Griffin A, Mahesh A, Tiwari VK. Disruption of the gene regulatory programme in neurodevelopmental disorders. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2022; 1865:194860. [PMID: 36007842 DOI: 10.1016/j.bbagrm.2022.194860] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 06/15/2023]
Abstract
Cortical development consists of a series of synchronised events, including fate transition of cortical progenitors, neuronal migration, specification and connectivity. It is becoming clear that gene expression programs governing these events rely on the interplay between signalling molecules, transcription factors and epigenetic mechanisms. When genetic or environmental factors disrupt expression of genes involved in important brain development processes, neurodevelopmental disorders can occur. This review aims to highlight how recent advances in technologies have helped uncover and imitate the gene regulatory mechanisms commonly disrupted in neurodevelopmental disorders.
Collapse
Affiliation(s)
- Aoife Griffin
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queens University, Belfast BT9 7BL, United Kingdom
| | - Arun Mahesh
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queens University, Belfast BT9 7BL, United Kingdom
| | - Vijay K Tiwari
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queens University, Belfast BT9 7BL, United Kingdom.
| |
Collapse
|
22
|
Jackson V, Hermann J, Tynan CJ, Rolfe DJ, Corey RA, Duncan AL, Noriega M, Chu A, Kalli AC, Jones EY, Sansom MSP, Martin-Fernandez ML, Seiradake E, Chavent M. The guidance and adhesion protein FLRT2 dimerizes in cis via dual small-X 3-small transmembrane motifs. Structure 2022; 30:1354-1365.e5. [PMID: 35700726 DOI: 10.1016/j.str.2022.05.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 03/03/2022] [Accepted: 05/18/2022] [Indexed: 10/18/2022]
Abstract
Fibronectin Leucine-rich Repeat Transmembrane (FLRT 1-3) proteins are a family of broadly expressed single-spanning transmembrane receptors that play key roles in development. Their extracellular domains mediate homotypic cell-cell adhesion and heterotypic protein interactions with other receptors to regulate cell adhesion and guidance. These in trans FLRT interactions determine the formation of signaling complexes of varying complexity and function. Whether FLRTs also interact at the surface of the same cell, in cis, remains unknown. Here, molecular dynamics simulations reveal two dimerization motifs in the FLRT2 transmembrane helix. Single particle tracking experiments show that these Small-X3-Small motifs synergize with a third dimerization motif encoded in the extracellular domain to permit the cis association and co-diffusion patterns of FLRT2 receptors on cells. These results may point to a competitive switching mechanism between in cis and in trans interactions, which suggests that homotypic FLRT interaction mirrors the functionalities of classic adhesion molecules.
Collapse
Affiliation(s)
- Verity Jackson
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 5RJ, UK
| | - Julia Hermann
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 5RJ, UK
| | - Christopher J Tynan
- Central Laser Facility, Research Complex at Harwell, Science and Technology Facilities Council, Harwell Campus, Didcot, OX11 0FA, UK
| | - Daniel J Rolfe
- Central Laser Facility, Research Complex at Harwell, Science and Technology Facilities Council, Harwell Campus, Didcot, OX11 0FA, UK
| | - Robin A Corey
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 5RJ, UK
| | - Anna L Duncan
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 5RJ, UK
| | - Maxime Noriega
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, 205 route de Narbonne, 31400 Toulouse, France
| | - Amy Chu
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 5RJ, UK
| | - Antreas C Kalli
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine and Astbury Center for Structural Molecular Biology, University of Leeds, Leeds, LS2 9NL, UK
| | - E Yvonne Jones
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Mark S P Sansom
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 5RJ, UK
| | - Marisa L Martin-Fernandez
- Central Laser Facility, Research Complex at Harwell, Science and Technology Facilities Council, Harwell Campus, Didcot, OX11 0FA, UK.
| | - Elena Seiradake
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 5RJ, UK.
| | - Matthieu Chavent
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, 205 route de Narbonne, 31400 Toulouse, France.
| |
Collapse
|
23
|
Yang M, Li D, Jiang Z, Li C, Ji S, Sun J, Chang Y, Ruan S, Wang Z, Liang R, Dai X, Li B, Zhao H. TGF-β-induced FLRT3 attenuation is essential for cancer-associated fibroblast-mediated epithelial-mesenchymal transition in colorectal cancer. Mol Cancer Res 2022; 20:1247-1259. [PMID: 35560224 DOI: 10.1158/1541-7786.mcr-21-0924] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 04/04/2022] [Accepted: 05/11/2022] [Indexed: 12/24/2022]
Abstract
Cancer-associated fibroblasts (CAFs) constitute a major component of the tumor microenvironment. The effects of CAFs on the progression of colorectal cancer (CRC) remain controversial. In this study, we found the ectopic overexpression of Fibronectin leucine-rich transmembrane protein 3 (FLRT3) inhibited the process of Epithelial-mesenchymal transition (EMT), as well as the proliferation, migration, invasion, and promote apoptosis of CRC cells, whereas silencing FLRT3 expression resulted in the opposite phenomenon. FLRT3 downregulation was associated with a poor prognosis in CRC. Also, FLRT3 expression was significantly related to some clinicopathological factors, including T stage (p=0.037), N stage (p=0.042), and E-cadherin (p=0.002) level. Via univariate and multivariate analyses, M stage (p<0.0001), FLRT3 (p=0.044), and E-cadherin (p=0.003) were associated with overall survival and were independent prognostic factors for it. Mechanistically, CAFs secreted TGF-β, which downregulated FLRT3 expression by activating SMAD4 to promote aggressive phenotypes in CRC cells. Moreover, FLRT3 repressed tumorigenesis and lung metastasis, which could be reversed by LY2109761, a dual inhibitor of TGF-β receptor type I and II. Treatment with LY2109761 increased IFN-γ expression in CD8+ T cells and reduced the number of regulatory T cells in the tumor microenvironment. Taken together, we revealed the metastasis-suppressive function of FLRT3, which was attenuated during the CAFs-mediated activation of the TGF-β/SMAD4 signaling pathway to promote EMT in CRC. LY2109761 that significantly inhibited metastasis could be a new treatment option for advanced CRC. Implications: CAFs enhance CRC aggressiveness by reducing FLRT3 expression through activating TGF-β/SMAD4 signaling pathway. CAFs-targeted therapy and/or LY2109761 were promising treatments for CRC.
Collapse
Affiliation(s)
- Mengdi Yang
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, shanghai, China
| | - Dan Li
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyuan Jiang
- Shanghai Cancer Institute, Renji Hospital, Jiaotong University School of Medicine, Shanghai, China
| | - Changcan Li
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Suyuan Ji
- School of Life Sciences, Xiamen Univeristy, Xiamen, Fujian, China
| | - Jing Sun
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, shanghai, China
| | - Yujie Chang
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, shanghai, China
| | - Shunyi Ruan
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, shanghai, China
| | - Zhiyu Wang
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, shanghai, China
| | - Rui Liang
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xueyu Dai
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Li
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Zhao
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, shanghai, China
| |
Collapse
|
24
|
Smith SJ, Guillon E, Holley SA. The roles of inter-tissue adhesion in development and morphological evolution. J Cell Sci 2022; 135:275268. [PMID: 35522159 PMCID: PMC9264361 DOI: 10.1242/jcs.259579] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The study of how neighboring tissues physically interact with each other, inter-tissue adhesion, is an emerging field at the interface of cell biology, biophysics and developmental biology. Inter-tissue adhesion can be mediated by either cell-extracellular matrix adhesion or cell-cell adhesion, and both the mechanisms and consequences of inter-tissue adhesion have been studied in vivo in numerous vertebrate and invertebrate species. In this Review, we discuss recent progress in understanding the many functions of inter-tissue adhesion in development and evolution. Inter-tissue adhesion can couple the motion of adjacent tissues, be the source of mechanical resistance that constrains morphogenesis, and transmit tension required for normal development. Tissue-tissue adhesion can also create mechanical instability that leads to tissue folding or looping. Transient inter-tissue adhesion can facilitate tissue invasion, and weak tissue adhesion can generate friction that shapes and positions tissues within the embryo. Lastly, we review studies that reveal how inter-tissue adhesion contributes to the diversification of animal morphologies.
Collapse
Affiliation(s)
- Sarah Jacquelyn Smith
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | - Emilie Guillon
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | - Scott A Holley
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
25
|
Shinmyo Y, Hamabe-Horiike T, Saito K, Kawasaki H. Investigation of the Mechanisms Underlying the Development and Evolution of the Cerebral Cortex Using Gyrencephalic Ferrets. Front Cell Dev Biol 2022; 10:847159. [PMID: 35386196 PMCID: PMC8977464 DOI: 10.3389/fcell.2022.847159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
The mammalian cerebral cortex has changed significantly during evolution. As a result of the increase in the number of neurons and glial cells in the cerebral cortex, its size has markedly expanded. Moreover, folds, called gyri and sulci, appeared on its surface, and its neuronal circuits have become much more complicated. Although these changes during evolution are considered to have been crucial for the acquisition of higher brain functions, the mechanisms underlying the development and evolution of the cerebral cortex of mammals are still unclear. This is, at least partially, because it is difficult to investigate these mechanisms using mice only. Therefore, genetic manipulation techniques for the cerebral cortex of gyrencephalic carnivore ferrets were developed recently. Furthermore, gene knockout was achieved in the ferret cerebral cortex using the CRISPR/Cas9 system. These techniques enabled molecular investigations using the ferret cerebral cortex. In this review, we will summarize recent findings regarding the mechanisms underlying the development and evolution of the mammalian cerebral cortex, mainly focusing on research using ferrets.
Collapse
Affiliation(s)
- Yohei Shinmyo
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Toshihide Hamabe-Horiike
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Kengo Saito
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
26
|
Sakai D, Sugawara T, Kurokawa T, Murakami Y, Tomosugi M, Masuta H, Sakata-Haga H, Hatta T, Shoji H. Hif1α-dependent hypoxia signaling contributes to the survival of deep-layer neurons and cortex formation in a mouse model. Mol Brain 2022; 15:28. [PMID: 35361248 PMCID: PMC8973788 DOI: 10.1186/s13041-022-00911-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 03/16/2022] [Indexed: 11/25/2022] Open
Abstract
Hypoxia-inducible factor 1 α (Hif1α) plays a crucial role in brain development. To study the function of Hif1α in early brain development, we generated neuroepithelial cell-specific Hif1α-knockout mice. Hif1α-knockout mice died soon after birth; these mice exhibited an abnormal head shape, indicating the presence of brain defects. Morphological analysis revealed that Hif1α ablation reduced the overall size of the brain, especially affecting the telencephalon. Neuronal apoptosis predominantly occurred in deep-layer neurons, consequently the alignment of cortical layers was severely disorganized in Hif1α knockout mice. Furthermore, we demonstrated that Vegf signaling contributes to the survival of deep-layer neurons as a downstream effector of Hif1α-dependent hypoxia signaling. Taken together, our findings demonstrate that Hif1α plays a critical role in the early stages of telencephalon development.
Collapse
Affiliation(s)
- Daisuke Sakai
- Department of Biology, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa, 920-0293, Japan.
| | - Takeru Sugawara
- Department of Medical Life Systems, Doshisha University, Kyotanabe, Kyoto, 610-0394, Japan
| | - Tomonori Kurokawa
- Department of Medical Life Systems, Doshisha University, Kyotanabe, Kyoto, 610-0394, Japan
| | - Yuki Murakami
- Department of Hygiene and Public Health, Kansai Medical University, Osaka, Hirakata, 573-1010, Japan
| | - Mitsuhiro Tomosugi
- Department of Anatomy, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan
| | - Hiroko Masuta
- Department of Anatomy, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan
| | - Hiromi Sakata-Haga
- Department of Anatomy, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan
| | - Toshihisa Hatta
- Department of Anatomy, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan
| | - Hiroki Shoji
- Department of Biology, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa, 920-0293, Japan
| |
Collapse
|
27
|
Shinmyo Y, Saito K, Hamabe-Horiike T, Kameya N, Ando A, Kawasaki K, Duong TAD, Sakashita M, Roboon J, Hattori T, Kannon T, Hosomichi K, Slezak M, Holt MG, Tajima A, Hori O, Kawasaki H. Localized astrogenesis regulates gyrification of the cerebral cortex. SCIENCE ADVANCES 2022; 8:eabi5209. [PMID: 35275722 PMCID: PMC8916738 DOI: 10.1126/sciadv.abi5209] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 01/19/2022] [Indexed: 06/14/2023]
Abstract
The development and evolution of mammalian higher cognition are represented by gyrification of the laminar cerebral cortex and astrocyte development, but their mechanisms and interrelationships remain unknown. Here, we show that localized astrogenesis plays an important role in gyri formation in the gyrencephalic cerebral cortex. In functional genetic experiments, we show that reducing astrocyte number prevents gyri formation in the ferret cortex, while increasing astrocyte number in mice, which do not have cortical folds, can induce gyrus-like protrusions. Morphometric analyses demonstrate that the vertical expansion of deep pallial regions achieved by localized astrogenesis is crucial for gyri formation. Furthermore, our findings suggest that localized astrogenesis by a positive feedback loop of FGF signaling is an important mechanism underlying cortical folding in gyrencephalic mammalian brains. Our findings reveal both the cellular mechanisms and the mechanical principle of gyrification in the mammalian brain.
Collapse
Affiliation(s)
- Yohei Shinmyo
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Kengo Saito
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Toshihide Hamabe-Horiike
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Narufumi Kameya
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Akitaka Ando
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Kanji Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Tung Anh Dinh Duong
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Masataka Sakashita
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Jureepon Roboon
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Tsuyoshi Hattori
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Takayuki Kannon
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Kazuyoshi Hosomichi
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Michal Slezak
- VIB Center for Brain and Disease Research, Herestraat 49, Leuven 3000, Belgium
- Łukasiewicz Research Network-PORT Polish Institute for Technology Development, 54-066 Wroclaw, Poland
| | - Matthew G. Holt
- VIB Center for Brain and Disease Research, Herestraat 49, Leuven 3000, Belgium
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal
| | - Atsushi Tajima
- Department of Bioinformatics and Genomics, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Osamu Hori
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| | - Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan
| |
Collapse
|
28
|
Convergent selective signaling impairment exposes the pathogenicity of latrophilin-3 missense variants linked to inheritable ADHD susceptibility. Mol Psychiatry 2022; 27:2425-2438. [PMID: 35393556 PMCID: PMC9135631 DOI: 10.1038/s41380-022-01537-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 03/09/2022] [Accepted: 03/21/2022] [Indexed: 12/13/2022]
Abstract
Latrophilin-3 (Lphn3; also known as ADGRL3) is a member of the adhesion G Protein Coupled Receptor subfamily, which participates in the stabilization and maintenance of neuronal networks by mediating intercellular adhesion through heterophilic interactions with transmembrane ligands. Polymorphisms modifying the Lphn3 gene are associated with attention-deficit/hyperactivity disorder (ADHD) in children and its persistence into adulthood. How these genetic alterations affect receptor function remains unknown. Here, we conducted the functional validation of distinct ADHD-related Lphn3 variants bearing mutations in the receptor's adhesion motif-containing extracellular region. We found that all variants tested disrupted the ability of Lphn3 to stabilize intercellular adhesion in a manner that was distinct between ligands classes, but which did not depend on ligand-receptor interaction parameters, thus pointing to altered intrinsic receptor signaling properties. Using G protein signaling biosensors, we determined that Lphn3 couples to Gαi1, Gαi2, Gαs, Gαq, and Gα13. However, all ADHD-related receptor variants consistently lacked intrinsic as well as ligand-dependent Gα13 coupling efficiency while maintaining unaltered coupling to Gαi, Gαs, and Gαq. Consistent with these alterations, actin remodeling functions as well as actin-relevant RhoA signaling normally displayed by the constitutively active Lphn3 receptor were impeded by select receptor variants, thus supporting additional signaling defects. Taken together, our data point to Gα13 selective signaling impairments as representing a disease-relevant pathogenicity pathway that can be inherited through Lphn3 gene polymorphisms. This study highlights the intricate interplay between Lphn3 GPCR functions and the actin cytoskeleton in modulating neurodevelopmental cues related to ADHD etiology.
Collapse
|
29
|
Ruff T, Peters C, Matsumoto A, Ihle SJ, Morales PA, Gaitanos L, Yonehara K, Del Toro D, Klein R. FLRT3 Marks Direction-Selective Retinal Ganglion Cells That Project to the Medial Terminal Nucleus. Front Mol Neurosci 2021; 14:790466. [PMID: 34955746 PMCID: PMC8696037 DOI: 10.3389/fnmol.2021.790466] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/19/2021] [Indexed: 11/23/2022] Open
Abstract
The mammalian retina extracts a multitude of diverse features from the visual scene such as color, contrast, and direction of motion. These features are transmitted separately to the brain by more than 40 different retinal ganglion cell (RGC) subtypes. However, so far only a few genetic markers exist to fully characterize the different RGC subtypes. Here, we present a novel genetic Flrt3-CreERT2 knock-in mouse that labels a small subpopulation of RGCs. Using single-cell injection of fluorescent dyes in Flrt3 positive RGCs, we distinguished four morphological RGC subtypes. Anterograde tracings using a fluorescent Cre-dependent Adeno-associated virus (AAV) revealed that a subgroup of Flrt3 positive RGCs specifically project to the medial terminal nucleus (MTN), which is part of the accessory optic system (AOS) and is essential in driving reflex eye movements for retinal image stabilization. Functional characterization using ex vivo patch-clamp recordings showed that the MTN-projecting Flrt3 RGCs preferentially respond to downward motion in an ON-fashion. These neurons distribute in a regular pattern and most of them are bistratified at the level of the ON and OFF bands of cholinergic starburst amacrine cells where they express the known ON-OFF direction-selective RGC marker CART. Together, our results indicate that MTN-projecting Flrt3 RGCs represent a new functionally homogeneous AOS projecting direction-selective RGC subpopulation.
Collapse
Affiliation(s)
- Tobias Ruff
- Department of Molecules, Signaling, and Development, Max Planck Institute of Neurobiology, Martinsried, Germany.,Laboratory of Biosensors and Bioelectronics, ETH Zürich, Zurich, Switzerland
| | - Christian Peters
- Department of Molecules, Signaling, and Development, Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Akihiro Matsumoto
- Department of Biomedicine, Nordic-EMBL Partnership for Molecular Medicine, Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Stephan J Ihle
- Laboratory of Biosensors and Bioelectronics, ETH Zürich, Zurich, Switzerland
| | - Pilar Alcalá Morales
- Department of Molecules, Signaling, and Development, Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Louise Gaitanos
- Department of Molecules, Signaling, and Development, Max Planck Institute of Neurobiology, Martinsried, Germany
| | - Keisuke Yonehara
- Department of Biomedicine, Nordic-EMBL Partnership for Molecular Medicine, Danish Research Institute of Translational Neuroscience - DANDRITE, Aarhus University, Aarhus, Denmark
| | - Daniel Del Toro
- Department of Molecules, Signaling, and Development, Max Planck Institute of Neurobiology, Martinsried, Germany.,Department of Biological Sciences, Faculty of Medicine, Institute of Neurosciences, IDIBAPS, CIBERNED, University of Barcelona, Barcelona, Spain
| | - Rüdiger Klein
- Department of Molecules, Signaling, and Development, Max Planck Institute of Neurobiology, Martinsried, Germany
| |
Collapse
|
30
|
Kyrousi C, O’Neill AC, Brazovskaja A, He Z, Kielkowski P, Coquand L, Di Giaimo R, D’ Andrea P, Belka A, Forero Echeverry A, Mei D, Lenge M, Cruceanu C, Buchsbaum IY, Khattak S, Fabien G, Binder E, Elmslie F, Guerrini R, Baffet AD, Sieber SA, Treutlein B, Robertson SP, Cappello S. Extracellular LGALS3BP regulates neural progenitor position and relates to human cortical complexity. Nat Commun 2021; 12:6298. [PMID: 34728600 PMCID: PMC8564519 DOI: 10.1038/s41467-021-26447-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 09/26/2021] [Indexed: 12/15/2022] Open
Abstract
Basal progenitors (BPs), including intermediate progenitors and basal radial glia, are generated from apical radial glia and are enriched in gyrencephalic species like humans, contributing to neuronal expansion. Shortly after generation, BPs delaminate towards the subventricular zone, where they further proliferate before differentiation. Gene expression alterations involved in BP delamination and function in humans are poorly understood. Here, we study the role of LGALS3BP, so far known as a cancer biomarker, which is a secreted protein enriched in human neural progenitors (NPCs). We show that individuals with LGALS3BP de novo variants exhibit altered local gyrification, sulcal depth, surface area and thickness in their cortex. Additionally, using cerebral organoids, human fetal tissues and mice, we show that LGALS3BP regulates the position of NPCs. Single-cell RNA-sequencing and proteomics reveal that LGALS3BP-mediated mechanisms involve the extracellular matrix in NPCs' anchoring and migration within the human brain. We propose that its temporal expression influences NPCs' delamination, corticogenesis and gyrification extrinsically.
Collapse
Affiliation(s)
- Christina Kyrousi
- grid.419548.50000 0000 9497 5095Max Planck Institute of Psychiatry, 80804 Munich, Germany ,grid.5216.00000 0001 2155 0800Present Address: First Department of Psychiatry, Medical School, National and Kapodistrian University of Athens, Greece and University Mental Health, Neurosciences and Precision Medicine Research Institute “Costas Stefanis”, Athens, Greece
| | - Adam C. O’Neill
- grid.29980.3a0000 0004 1936 7830Department of Women’s and Children’s Health, University of Otago, 9054 Dunedin, New Zealand
| | - Agnieska Brazovskaja
- grid.419518.00000 0001 2159 1813Max Planck Institute for Evolutionary Anthropology, 04103 Leipzig, Germany
| | - Zhisong He
- grid.419518.00000 0001 2159 1813Max Planck Institute for Evolutionary Anthropology, 04103 Leipzig, Germany ,grid.5801.c0000 0001 2156 2780ETH Zurich, Department of Biosystems Science and Engineering, 4058 Basel, Switzerland
| | - Pavel Kielkowski
- grid.6936.a0000000123222966Department of Chemistry, Chair of Organic Chemistry II, Center for Integrated Protein Science (CIPSM), Technische Universität München, Garching, Germany ,grid.5252.00000 0004 1936 973XPresent Address: Department Chemie Ludwig-Maximilians-Universität München Butenandtstr. 5-13, 81377 München, Germany
| | - Laure Coquand
- grid.4444.00000 0001 2112 9282Institut Curie, PSL Research University, CNRS, UMR 144, 26 rue d’Ulm, F-75005 Paris, France
| | - Rossella Di Giaimo
- grid.419548.50000 0000 9497 5095Max Planck Institute of Psychiatry, 80804 Munich, Germany ,grid.4691.a0000 0001 0790 385XDepartment of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Pierpaolo D’ Andrea
- grid.419548.50000 0000 9497 5095Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Alexander Belka
- grid.419548.50000 0000 9497 5095Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | | | - Davide Mei
- grid.413181.e0000 0004 1757 8562Neuroscience Department, Children’s Hospital A. Meyer-University of Florence, 50139 Florence, Italy
| | - Matteo Lenge
- grid.413181.e0000 0004 1757 8562Neuroscience Department, Children’s Hospital A. Meyer-University of Florence, 50139 Florence, Italy
| | - Cristiana Cruceanu
- grid.419548.50000 0000 9497 5095Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Isabel Y. Buchsbaum
- grid.419548.50000 0000 9497 5095Max Planck Institute of Psychiatry, 80804 Munich, Germany ,grid.5252.00000 0004 1936 973XGraduate School of Systemic Neurosciences, Ludwig-Maximilians-University, 82152 Munich Planegg, Germany
| | - Shahryar Khattak
- grid.4488.00000 0001 2111 7257DFG-Research Center and Cluster of Excellence for Regenerative Therapies (CRTD), School of Medicine, Technical University Dresden, 01307 Dresden, Germany ,grid.4912.e0000 0004 0488 7120Present Address: Royal College of Surgeons Ireland (RCSI) in Bahrain, Adliya, Kingdom of Bahrain
| | - Guimiot Fabien
- grid.50550.350000 0001 2175 4109Unité de Foetopathologie, Assistance Publique-Hôpitaux de Paris, CHU Robert Debré, F-75019 Paris, France
| | - Elisabeth Binder
- grid.419548.50000 0000 9497 5095Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Frances Elmslie
- grid.4464.20000 0001 2161 2573South West Thames Regional Genetics Service, St George’s, University of London, London, SW17 0RE UK
| | - Renzo Guerrini
- grid.413181.e0000 0004 1757 8562Neuroscience Department, Children’s Hospital A. Meyer-University of Florence, 50139 Florence, Italy
| | - Alexandre D. Baffet
- grid.4444.00000 0001 2112 9282Institut Curie, PSL Research University, CNRS, UMR 144, 26 rue d’Ulm, F-75005 Paris, France
| | - Stephan A. Sieber
- grid.6936.a0000000123222966Department of Chemistry, Chair of Organic Chemistry II, Center for Integrated Protein Science (CIPSM), Technische Universität München, Garching, Germany
| | - Barbara Treutlein
- grid.419518.00000 0001 2159 1813Max Planck Institute for Evolutionary Anthropology, 04103 Leipzig, Germany ,grid.5801.c0000 0001 2156 2780ETH Zurich, Department of Biosystems Science and Engineering, 4058 Basel, Switzerland
| | - Stephen P. Robertson
- grid.29980.3a0000 0004 1936 7830Department of Women’s and Children’s Health, University of Otago, 9054 Dunedin, New Zealand
| | - Silvia Cappello
- grid.419548.50000 0000 9497 5095Max Planck Institute of Psychiatry, 80804 Munich, Germany
| |
Collapse
|
31
|
Transcription factor 4 controls positioning of cortical projection neurons through regulation of cell adhesion. Mol Psychiatry 2021; 26:6562-6577. [PMID: 33963287 DOI: 10.1038/s41380-021-01119-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 04/02/2021] [Accepted: 04/13/2021] [Indexed: 02/07/2023]
Abstract
The establishment of neural circuits depends on precise neuronal positioning in the cortex, which occurs via a tightly coordinated process of neuronal differentiation, migration, and terminal localization. Deficits in this process have been implicated in several psychiatric disorders. Here, we show that the transcription factor Tcf4 controls neuronal positioning during brain development. Tcf4-deficient neurons become mispositioned in clusters when their migration to the cortical plate is complete. We reveal that Tcf4 regulates the expression of cell adhesion molecules to control neuronal positioning. Furthermore, through in vivo extracellular electrophysiology, we show that neuronal functions are disrupted after the loss of Tcf4. TCF4 mutations are strongly associated with schizophrenia and cause Pitt-Hopkins syndrome, which is characterized by severe intellectual disability. Thus, our results not only reveal the importance of neuronal positioning in brain development but also provide new insights into the potential mechanisms underlying neurological defects linked to TCF4 mutations.
Collapse
|
32
|
Abstract
The human brain is characterized by the large size and intricate folding of its cerebral cortex, which are fundamental for our higher cognitive function and frequently altered in pathological dysfunction. Cortex folding is not unique to humans, nor even to primates, but is common across mammals. Cortical growth and folding are the result of complex developmental processes that involve neural stem and progenitor cells and their cellular lineages, the migration and differentiation of neurons, and the genetic programs that regulate and fine-tune these processes. All these factors combined generate mechanical stress and strain on the developing neural tissue, which ultimately drives orderly cortical deformation and folding. In this review we examine and summarize the current knowledge on the molecular, cellular, histogenic and mechanical mechanisms that are involved in and influence folding of the cerebral cortex, and how they emerged and changed during mammalian evolution. We discuss the main types of pathological malformations of human cortex folding, their specific developmental origin, and how investigating their genetic causes has illuminated our understanding of key events involved. We close our review by presenting the state-of-the-art animal and in vitro models of cortex folding that are currently used to study these devastating developmental brain disorders in children, and what are the main challenges that remain ahead of us to fully understand brain folding.
Collapse
Affiliation(s)
- Lucia Del Valle Anton
- Instituto de Neurociencias, Agencia Estatal Consejo Superior de Investigaciones Científicas, San Juan de Alicante, Alicante, Spain
| | - Victor Borrell
- Instituto de Neurociencias, Agencia Estatal Consejo Superior de Investigaciones Científicas, San Juan de Alicante, Alicante, Spain
| |
Collapse
|
33
|
He CH, Zhang L, Song NN, Mei WY, Chen JY, Hu L, Zhang Q, Wang YB, Ding YQ. Satb2 Regulates EphA7 to Control Soma Spacing and Self-Avoidance of Cortical Pyramidal Neurons. Cereb Cortex 2021; 32:2321-2331. [PMID: 34546353 DOI: 10.1093/cercor/bhab321] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Soma spacing and dendritic arborization during brain development are key events for the establishment of proper neural circuitry and function. Transcription factor Satb2 is a molecular node in regulating the development of the cerebral cortex, as shown by the facts that Satb2 is required for the regionalization of retrosplenial cortex, the determination of callosal neuron fate, and the regulation of soma spacing and dendritic self-avoidance of cortical pyramidal neurons. In this study, we explored downstream effectors that mediate the Satb2-implicated soma spacing and dendritic self-avoidance. First, RNA-seq analysis of the cortex revealed differentially expressed genes between control and Satb2 CKO mice. Among them, EphA7 transcription was dramatically increased in layers II/III of Satb2 CKO cortex. Overexpression of EphA7 in the late-born cortical neurons of wild-type mice via in utero electroporation resulted in soma clumping and impaired self-avoidance of affected pyramidal neurons, which resembles the phenotypes caused by knockdown of Satb2 expression. Importantly, the phenotypes by Satb2 knockdown was rescued by reducing EphA7 expression in the cortex. Finally, ChIP and luciferase reporter assays indicated a direct suppression of EphA7 expression by Satb2. These findings provide new insights into the complexity of transcriptional regulation of the morphogenesis of cerebral cortex.
Collapse
Affiliation(s)
- Chun-Hui He
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Lei Zhang
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Ning-Ning Song
- Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China.,State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Wan-Ying Mei
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Jia-Yin Chen
- Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China.,State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Ling Hu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Qiong Zhang
- Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China.,State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Yu-Bing Wang
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai 200092, China
| | - Yu-Qiang Ding
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai 200120, China.,Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai 200092, China.,Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China.,State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China
| |
Collapse
|
34
|
Libé-Philippot B, Vanderhaeghen P. Cellular and Molecular Mechanisms Linking Human Cortical Development and Evolution. Annu Rev Genet 2021; 55:555-581. [PMID: 34535062 DOI: 10.1146/annurev-genet-071719-020705] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The cerebral cortex is at the core of brain functions that are thought to be particularly developed in the human species. Human cortex specificities stem from divergent features of corticogenesis, leading to increased cortical size and complexity. Underlying cellular mechanisms include prolonged patterns of neuronal generation and maturation, as well as the amplification of specific types of stem/progenitor cells. While the gene regulatory networks of corticogenesis appear to be largely conserved among all mammals including humans, they have evolved in primates, particularly in the human species, through the emergence of rapidly divergent transcriptional regulatory elements, as well as recently duplicated novel genes. These human-specific molecular features together control key cellular milestones of human corticogenesis and are often affected in neurodevelopmental disorders, thus linking human neural development, evolution, and diseases. Expected final online publication date for the Annual Review of Genetics, Volume 55 is November 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Baptiste Libé-Philippot
- VIB-KU Leuven Center for Brain & Disease Research, KU Leuven Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium; .,Institut de Recherches Interdisciplinaires en Biologie Humaine et Moléculaire (IRIBHM) and ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Pierre Vanderhaeghen
- VIB-KU Leuven Center for Brain & Disease Research, KU Leuven Department of Neurosciences, Leuven Brain Institute, 3000 Leuven, Belgium; .,Institut de Recherches Interdisciplinaires en Biologie Humaine et Moléculaire (IRIBHM) and ULB Neuroscience Institute (UNI), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| |
Collapse
|
35
|
Kerimoglu C, Pham L, Tonchev AB, Sakib MS, Xie Y, Sokpor G, Ulmke PA, Kaurani L, Abbas E, Nguyen H, Rosenbusch J, Michurina A, Capece V, Angelova M, Maricic N, Brand-Saberi B, Esgleas M, Albert M, Minkov R, Kovachev E, Teichmann U, Seong RH, Huttner WB, Nguyen HP, Stoykova A, Staiger JF, Fischer A, Tuoc T. H3 acetylation selectively promotes basal progenitor proliferation and neocortex expansion. SCIENCE ADVANCES 2021; 7:eabc6792. [PMID: 34524839 PMCID: PMC8443185 DOI: 10.1126/sciadv.abc6792] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/23/2021] [Indexed: 06/13/2023]
Abstract
Increase in the size of human neocortex―acquired in evolution―accounts for the unique cognitive capacity of humans. This expansion reflects the evolutionarily enhanced proliferative ability of basal progenitors (BPs), including the basal radial glia and basal intermediate progenitors (bIPs) in mammalian cortex, which may have been acquired through epigenetic alterations in BPs. However, how the epigenome in BPs differs across species is not known. Here, we report that histone H3 acetylation is a key epigenetic regulation in bIP amplification and cortical expansion. Through epigenetic profiling of sorted bIPs, we show that histone H3 lysine 9 acetylation (H3K9ac) is low in murine bIPs and high in human bIPs. Elevated H3K9ac preferentially increases bIP proliferation, increasing the size and folding of the normally smooth mouse neocortex. H3K9ac drives bIP amplification by increasing expression of the evolutionarily regulated gene, Trnp1, in developing cortex. Our findings demonstrate a previously unknown mechanism that controls cortical architecture.
Collapse
Affiliation(s)
- Cemil Kerimoglu
- German Center for Neurodegenerative Diseases, 37077 Goettingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
| | - Linh Pham
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Anton B. Tonchev
- Max Planck Institute for Biophysical Chemistry, 37077 Goettingen, Germany
- Departments of Anatomy and Cell Biology and Stem Cell Biology, Research Institute, Medical University of Varna, Varna 9002, Bulgaria
| | - M. Sadman Sakib
- German Center for Neurodegenerative Diseases, 37077 Goettingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
| | - Yuanbin Xie
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Gannan Medical University, Ganzhou 341000, The People’s Republic of China
| | - Godwin Sokpor
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Pauline Antonie Ulmke
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
| | - Lalit Kaurani
- German Center for Neurodegenerative Diseases, 37077 Goettingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
| | - Eman Abbas
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
- Zoology Department, Faculty of Science, Alexandria University, Alexandria, Egypt
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, 01307 Dresden, Germany
| | - Huong Nguyen
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
- Faculty of Biotechnology, Thai Nguyen University of Sciences, Thai Nguyen, Vietnam
| | - Joachim Rosenbusch
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
| | | | - Vincenzo Capece
- German Center for Neurodegenerative Diseases, 37077 Goettingen, Germany
| | - Meglena Angelova
- Departments of Anatomy and Cell Biology and Stem Cell Biology, Research Institute, Medical University of Varna, Varna 9002, Bulgaria
| | - Nenad Maricic
- Institute of Anatomy and Molecular Embryology, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Beate Brand-Saberi
- Institute of Anatomy and Molecular Embryology, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Miriam Esgleas
- Institute of Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health, Munich, Germany
| | - Mareike Albert
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, 01307 Dresden, Germany
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Radoslav Minkov
- Specialized Hospital for Obstetrics and Gynecology “Prof. Dimitar Stamatov” –Varna, Medical University of Varna, Varna 9002, Bulgaria
| | - Emil Kovachev
- Specialized Hospital for Obstetrics and Gynecology “Prof. Dimitar Stamatov” –Varna, Medical University of Varna, Varna 9002, Bulgaria
| | - Ulrike Teichmann
- Max Planck Institute for Biophysical Chemistry, 37077 Goettingen, Germany
| | - Rho H. Seong
- School of Biological Sciences and Institute for Molecular Biology and Genetics, Seoul National University, Seoul, South Korea
| | - Wieland B. Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Huu Phuc Nguyen
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
| | - Anastassia Stoykova
- Max Planck Institute for Biophysical Chemistry, 37077 Goettingen, Germany
- DFG Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), 37075 Goettingen, Germany
| | - Jochen F. Staiger
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
- DFG Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), 37075 Goettingen, Germany
| | - Andre Fischer
- German Center for Neurodegenerative Diseases, 37077 Goettingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
- DFG Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), 37075 Goettingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells” (MBExC), Georg-August-University Goettingen, 37075 Goettingen, Germany
| | - Tran Tuoc
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, 37075 Goettingen, Germany
- Department of Human Genetics, Ruhr University of Bochum, 44791 Bochum, Germany
- DFG Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), 37075 Goettingen, Germany
| |
Collapse
|
36
|
Han S, Okawa S, Wilkinson GA, Ghazale H, Adnani L, Dixit R, Tavares L, Faisal I, Brooks MJ, Cortay V, Zinyk D, Sivitilli A, Li S, Malik F, Ilnytskyy Y, Angarica VE, Gao J, Chinchalongporn V, Oproescu AM, Vasan L, Touahri Y, David LA, Raharjo E, Kim JW, Wu W, Rahmani W, Chan JAW, Kovalchuk I, Attisano L, Kurrasch D, Dehay C, Swaroop A, Castro DS, Biernaskie J, Del Sol A, Schuurmans C. Proneural genes define ground-state rules to regulate neurogenic patterning and cortical folding. Neuron 2021; 109:2847-2863.e11. [PMID: 34407390 DOI: 10.1016/j.neuron.2021.07.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 05/19/2021] [Accepted: 07/08/2021] [Indexed: 02/06/2023]
Abstract
Asymmetric neuronal expansion is thought to drive evolutionary transitions between lissencephalic and gyrencephalic cerebral cortices. We report that Neurog2 and Ascl1 proneural genes together sustain neurogenic continuity and lissencephaly in rodent cortices. Using transgenic reporter mice and human cerebral organoids, we found that Neurog2 and Ascl1 expression defines a continuum of four lineage-biased neural progenitor cell (NPC) pools. Double+ NPCs, at the hierarchical apex, are least lineage restricted due to Neurog2-Ascl1 cross-repression and display unique features of multipotency (more open chromatin, complex gene regulatory network, G2 pausing). Strikingly, selectively eliminating double+ NPCs by crossing Neurog2-Ascl1 split-Cre mice with diphtheria toxin-dependent "deleter" strains locally disrupts Notch signaling, perturbs neurogenic symmetry, and triggers cortical folding. In support of our discovery that double+ NPCs are Notch-ligand-expressing "niche" cells that control neurogenic periodicity and cortical folding, NEUROG2, ASCL1, and HES1 transcript distribution is modular (adjacent high/low zones) in gyrencephalic macaque cortices, prefiguring future folds.
Collapse
Affiliation(s)
- Sisu Han
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Satoshi Okawa
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg; Integrated BioBank of Luxembourg, 3555, 3531 Dudelange, Luxembourg
| | - Grey Atteridge Wilkinson
- Department of Biochemistry and Molecular Biology, ACHRI, HBI, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Hussein Ghazale
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Lata Adnani
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada; Department of Biochemistry and Molecular Biology, ACHRI, HBI, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Rajiv Dixit
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ligia Tavares
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Imrul Faisal
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Matthew J Brooks
- Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892-1204, USA
| | - Veronique Cortay
- Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France
| | - Dawn Zinyk
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada
| | - Adam Sivitilli
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada; Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Saiqun Li
- Department of Biochemistry and Molecular Biology, ACHRI, HBI, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Faizan Malik
- Department of Medical Genetics, ACHRI, HBI, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Yaroslav Ilnytskyy
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Vladimir Espinosa Angarica
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg
| | - Jinghua Gao
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Vorapin Chinchalongporn
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ana-Maria Oproescu
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Lakshmy Vasan
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Yacine Touahri
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Luke Ajay David
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Eko Raharjo
- Department of Comparative Biology and Experimental Medicine, HBI, ACHRI, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Jung-Woong Kim
- Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892-1204, USA
| | - Wei Wu
- Department of Pathology and Laboratory Medicine, Charbonneau Cancer Institute, HBI, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Waleed Rahmani
- Department of Comparative Biology and Experimental Medicine, HBI, ACHRI, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Jennifer Ai-Wen Chan
- Department of Pathology and Laboratory Medicine, Charbonneau Cancer Institute, HBI, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Igor Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Liliana Attisano
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada; Donnelly Centre, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Deborah Kurrasch
- Department of Medical Genetics, ACHRI, HBI, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Colette Dehay
- Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, 69500 Bron, France
| | - Anand Swaroop
- Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892-1204, USA
| | - Diogo S Castro
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Jeff Biernaskie
- Department of Comparative Biology and Experimental Medicine, HBI, ACHRI, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Antonio Del Sol
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4362 Esch-sur-Alzette, Luxembourg; CIC bioGUNE, Bizkaia Technology Park, 48160 Derio, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao 48013, Spain
| | - Carol Schuurmans
- Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, ON M4N 3M5, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Biochemistry and Molecular Biology, ACHRI, HBI, University of Calgary, Calgary, AB T2N 4N1, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
37
|
Fleitas C, Marfull-Oromí P, Chauhan D, Del Toro D, Peguera B, Zammou B, Rocandio D, Klein R, Espinet C, Egea J. FLRT2 and FLRT3 Cooperate in Maintaining the Tangential Migratory Streams of Cortical Interneurons during Development. J Neurosci 2021; 41:7350-7362. [PMID: 34301831 PMCID: PMC8412983 DOI: 10.1523/jneurosci.0380-20.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/29/2021] [Accepted: 07/06/2021] [Indexed: 02/08/2023] Open
Abstract
Neuron migration is a hallmark of nervous system development that allows gathering of neurons from different origins for assembling of functional neuronal circuits. Cortical inhibitory interneurons arise in the ventral telencephalon and migrate tangentially forming three transient migratory streams in the cortex before reaching the final laminar destination. Although migration defects lead to the disruption of inhibitory circuits and are linked to aspects of psychiatric disorders such as autism and schizophrenia, the molecular mechanisms controlling cortical interneuron development and final layer positioning are incompletely understood. Here, we show that mouse embryos with a double deletion of FLRT2 and FLRT3 genes encoding cell adhesion molecules exhibit an abnormal distribution of interneurons within the streams during development, which in turn, affect the layering of somatostatin+ interneurons postnatally. Mechanistically, FLRT2 and FLRT3 proteins act in a noncell-autonomous manner, possibly through a repulsive mechanism. In support of such a conclusion, double knockouts deficient in the repulsive receptors for FLRTs, Unc5B and Unc5D, also display interneuron defects during development, similar to the FLRT2/FLRT3 mutants. Moreover, FLRT proteins are chemorepellent ligands for developing interneurons in vitro, an effect that is in part dependent on FLRT-Unc5 interaction. Together, we propose that FLRTs act through Unc5 receptors to control cortical interneuron distribution in a mechanism that involves cell repulsion.SIGNIFICANCE STATEMENT Disruption of inhibitory cortical circuits is responsible for some aspects of psychiatric disorders such as schizophrenia or autism. These defects include interneuron migration during development. A crucial step during this process is the formation of three transient migratory streams within the developing cortex that determine the timing of interneuron final positioning and the formation of functional cortical circuits in the adult. We report that FLRT proteins are required for the proper distribution of interneurons within the cortical migratory streams and for the final laminar allocation in the postnatal cortex. These results expand the multifunctional role of FLRTs during nervous system development in addition to the role of FLRTs in axon guidance and the migration of excitatory cortical neurons.
Collapse
Affiliation(s)
- Catherine Fleitas
- Lleida Biomedical Research Institute, University of Lleida, Lleida 25198, Spain
| | - Pau Marfull-Oromí
- Lleida Biomedical Research Institute, University of Lleida, Lleida 25198, Spain
| | - Disha Chauhan
- Lleida Biomedical Research Institute, University of Lleida, Lleida 25198, Spain
| | - Daniel Del Toro
- Max Planck Institute of Neurobiology, 82152 Martinsried, Germany
| | - Blanca Peguera
- Lleida Biomedical Research Institute, University of Lleida, Lleida 25198, Spain
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences, University of Frankfurt, D-60438 Frankfurt am Main, Germany
| | - Bahira Zammou
- Lleida Biomedical Research Institute, University of Lleida, Lleida 25198, Spain
| | - Daniel Rocandio
- Lleida Biomedical Research Institute, University of Lleida, Lleida 25198, Spain
| | - Rüdiger Klein
- Max Planck Institute of Neurobiology, 82152 Martinsried, Germany
| | - Carme Espinet
- Lleida Biomedical Research Institute, University of Lleida, Lleida 25198, Spain
| | - Joaquim Egea
- Lleida Biomedical Research Institute, University of Lleida, Lleida 25198, Spain
- Serra Hunter Associate Professor, Government of Catalonia, 08007, Spain
| |
Collapse
|
38
|
Hickmott RA, Bosakhar A, Quezada S, Barresi M, Walker DW, Ryan AL, Quigley A, Tolcos M. The One-Stop Gyrification Station - Challenges and New Technologies. Prog Neurobiol 2021; 204:102111. [PMID: 34166774 DOI: 10.1016/j.pneurobio.2021.102111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/31/2021] [Accepted: 06/18/2021] [Indexed: 12/12/2022]
Abstract
The evolution of the folded cortical surface is an iconic feature of the human brain shared by a subset of mammals and considered pivotal for the emergence of higher-order cognitive functions. While our understanding of the neurodevelopmental processes involved in corticogenesis has greatly advanced over the past 70 years of brain research, the fundamental mechanisms that result in gyrification, along with its originating cytoarchitectural location, remain largely unknown. This review brings together numerous approaches to this basic neurodevelopmental problem, constructing a narrative of how various models, techniques and tools have been applied to the study of gyrification thus far. After a brief discussion of core concepts and challenges within the field, we provide an analysis of the significant discoveries derived from the parallel use of model organisms such as the mouse, ferret, sheep and non-human primates, particularly with regard to how they have shaped our understanding of cortical folding. We then focus on the latest developments in the field and the complementary application of newly emerging technologies, such as cerebral organoids, advanced neuroimaging techniques, and atomic force microscopy. Particular emphasis is placed upon the use of novel computational and physical models in regard to the interplay of biological and physical forces in cortical folding.
Collapse
Affiliation(s)
- Ryan A Hickmott
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, 3083, Australia; BioFab3D@ACMD, St Vincent's Hospital Melbourne, Fitzroy, VIC, 3065, Australia
| | - Abdulhameed Bosakhar
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, 3083, Australia
| | - Sebastian Quezada
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, 3083, Australia
| | - Mikaela Barresi
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, 3083, Australia
| | - David W Walker
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, 3083, Australia
| | - Amy L Ryan
- Hastings Centre for Pulmonary Research, Department of Pulmonary, Critical Care and Sleep Medicine, USC Keck School of Medicine, University of Southern California, CA, USA and Department of Stem Cell and Regenerative Medicine, University of Southern California, CA, 90033, USA
| | - Anita Quigley
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, 3083, Australia; BioFab3D@ACMD, St Vincent's Hospital Melbourne, Fitzroy, VIC, 3065, Australia; School of Engineering, RMIT University, Melbourne, VIC, 3000, Australia; Department of Medicine, University of Melbourne, St Vincent's Hospital, Fitzroy, VIC, 3065, Australia; ARC Centre of Excellence in Electromaterials Science, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Mary Tolcos
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, 3083, Australia.
| |
Collapse
|
39
|
Zhu N, Zhang C, Prakash A, Hou Z, Liu W, She W, Morris A, Sik Kim K. Therapeutic development of group B Streptococcus meningitis by targeting a host cell signaling network involving EGFR. EMBO Mol Med 2021; 13:e12651. [PMID: 33474818 PMCID: PMC7933950 DOI: 10.15252/emmm.202012651] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
Group B Streptococcus (GBS) remains the most common Gram-positive bacterium causing neonatal meningitis and GBS meningitis continues to be an important cause of mortality and morbidity. In this study, we showed that GBS penetration into the brain occurred initially in the meningeal and cortex capillaries, and exploits a defined host cell signaling network comprised of S1P2 , EGFR, and CysLT1. GBS exploitation of such network in penetration of the blood-brain barrier was demonstrated by targeting S1P2 , EGFR, and CysLT1 using pharmacological inhibition, gene knockout and knockdown cells, and gene knockout animals, as well as interrogation of the network (up- and downstream of each other). More importantly, counteracting such targets as a therapeutic adjunct to antibiotic therapy was beneficial in improving the outcome of animals with GBS meningitis. These findings indicate that investigating GBS penetration of the blood-brain barrier provides a novel approach for therapeutic development of GBS meningitis.
Collapse
Affiliation(s)
- Ningyu Zhu
- Division of Pediatric Infectious DiseasesJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Chengxian Zhang
- Division of Pediatric Infectious DiseasesJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Atish Prakash
- Division of Pediatric Infectious DiseasesJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Zheng Hou
- Division of Pediatric Infectious DiseasesJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Wei Liu
- Division of Pediatric Infectious DiseasesJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Weifeng She
- Division of Pediatric Infectious DiseasesJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Andrew Morris
- Division of Cardiovascular MedicineThe Gill Heart InstituteUniversity of KentuckyLexingtonKYUSA
| | - Kwang Sik Kim
- Division of Pediatric Infectious DiseasesJohns Hopkins University School of MedicineBaltimoreMDUSA
| |
Collapse
|
40
|
Dong X, Yang L, Liu K, Ji X, Tang C, Li W, Ma L, Mei Y, Peng T, Feng B, Wu Z, Tang Q, Gao Y, Yan K, Zhou W, Xiong M. Transcriptional networks identify synaptotagmin-like 3 as a regulator of cortical neuronal migration during early neurodevelopment. Cell Rep 2021; 34:108802. [PMID: 33657377 DOI: 10.1016/j.celrep.2021.108802] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 12/25/2020] [Accepted: 02/08/2021] [Indexed: 02/07/2023] Open
Abstract
Human brain development is a complex process involving neural proliferation, differentiation, and migration that are directed by many essential cellular factors and drivers. Here, using the NetBID2 algorithm and developing human brain RNA sequencing dataset, we identify synaptotagmin-like 3 (SYTL3) as one of the top drivers of early human brain development. Interestingly, SYTL3 exhibits high activity but low expression in both early developmental human cortex and human embryonic stem cell (hESC)-derived neurons. Knockout of SYTL3 (SYTL3-KO) in human neurons or knockdown of Sytl3 in embryonic mouse cortex markedly promotes neuronal migration. SYTL3-KO causes an abnormal distribution of deep-layer neurons in brain organoids and reduces presynaptic neurotransmitter release in hESC-derived neurons. We further demonstrate that SYTL3-KO-accelerated neuronal migration is modulated by high expression of matrix metalloproteinases. Together, based on bioinformatics and biological experiments, we identify SYTL3 as a regulator of cortical neuronal migration in human and mouse developing brains.
Collapse
Affiliation(s)
- Xinran Dong
- Molecular Medical Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai 201102, China
| | - Lin Yang
- Molecular Medical Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai 201102, China
| | - Kaiyi Liu
- Molecular Medical Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai 201102, China
| | - Xiaoli Ji
- Molecular Medical Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai 201102, China; Stem Cell Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Chuanqing Tang
- Stem Cell Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Wanxing Li
- Molecular Medical Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai 201102, China
| | - Ling Ma
- Stem Cell Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Yuting Mei
- Stem Cell Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Ting Peng
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Ban Feng
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, Shanghai, China
| | - Ziyan Wu
- Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, Shanghai, China
| | - Qingyuan Tang
- Stem Cell Center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Yanyan Gao
- Ultrasonography Department, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Kai Yan
- Key Laboratory of Neonatal Diseases, Ministry of Health, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Wenhao Zhou
- Molecular Medical Center, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai 201102, China; Key Laboratory of Neonatal Diseases, Ministry of Health, Children's Hospital of Fudan University, Shanghai 201102, China.
| | - Man Xiong
- Stem Cell Center, Children's Hospital of Fudan University, Shanghai 201102, China.
| |
Collapse
|
41
|
Interneuron Origins in the Embryonic Porcine Medial Ganglionic Eminence. J Neurosci 2021; 41:3105-3119. [PMID: 33637558 DOI: 10.1523/jneurosci.2738-20.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/18/2020] [Accepted: 01/08/2021] [Indexed: 02/06/2023] Open
Abstract
Interneurons contribute to the complexity of neural circuits and maintenance of normal brain function. Rodent interneurons originate in embryonic ganglionic eminences, but developmental origins in other species are less understood. Here, we show that transcription factor expression patterns in porcine embryonic subpallium are similar to rodents, delineating a distinct medial ganglionic eminence (MGE) progenitor domain. On the basis of Nkx2.1, Lhx6, and Dlx2 expression, in vitro differentiation into neurons expressing GABA, and robust migratory capacity in explant assays, we propose that cortical and hippocampal interneurons originate from a porcine MGE region. Following xenotransplantation into adult male and female rat hippocampus, we further demonstrate that porcine MGE progenitors, like those from rodents, migrate and differentiate into morphologically distinct interneurons expressing GABA. Our findings reveal that basic rules for interneuron development are conserved across species, and that porcine embryonic MGE progenitors could serve as a valuable source for interneuron-based xenotransplantation therapies.SIGNIFICANCE STATEMENT Here we demonstrate that porcine medial ganglionic eminence, like rodents, exhibit a distinct transcriptional and interneuron-specific antibody profile, in vitro migratory capacity and are amenable to xenotransplantation. This is the first comprehensive examination of embryonic interneuron origins in the pig; and because a rich neurodevelopmental literature on embryonic mouse medial ganglionic eminence exists (with some additional characterizations in other species, e.g., monkey and human), our work allows direct neurodevelopmental comparisons with this literature.
Collapse
|
42
|
Oproescu AM, Han S, Schuurmans C. New Insights Into the Intricacies of Proneural Gene Regulation in the Embryonic and Adult Cerebral Cortex. Front Mol Neurosci 2021; 14:642016. [PMID: 33658912 PMCID: PMC7917194 DOI: 10.3389/fnmol.2021.642016] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 01/26/2021] [Indexed: 12/21/2022] Open
Abstract
Historically, the mammalian brain was thought to lack stem cells as no new neurons were found to be made in adulthood. That dogma changed ∼25 years ago with the identification of neural stem cells (NSCs) in the adult rodent forebrain. However, unlike rapidly self-renewing mature tissues (e.g., blood, intestinal crypts, skin), the majority of adult NSCs are quiescent, and those that become 'activated' are restricted to a few neurogenic zones that repopulate specific brain regions. Conversely, embryonic NSCs are actively proliferating and neurogenic. Investigations into the molecular control of the quiescence-to-proliferation-to-differentiation continuum in the embryonic and adult brain have identified proneural genes encoding basic-helix-loop-helix (bHLH) transcription factors (TFs) as critical regulators. These bHLH TFs initiate genetic programs that remove NSCs from quiescence and drive daughter neural progenitor cells (NPCs) to differentiate into specific neural cell subtypes, thereby contributing to the enormous cellular diversity of the adult brain. However, new insights have revealed that proneural gene activities are context-dependent and tightly regulated. Here we review how proneural bHLH TFs are regulated, with a focus on the murine cerebral cortex, drawing parallels where appropriate to other organisms and neural tissues. We discuss upstream regulatory events, post-translational modifications (phosphorylation, ubiquitinylation), protein-protein interactions, epigenetic and metabolic mechanisms that govern bHLH TF expression, stability, localization, and consequent transactivation of downstream target genes. These tight regulatory controls help to explain paradoxical findings of changes to bHLH activity in different cellular contexts.
Collapse
Affiliation(s)
- Ana-Maria Oproescu
- Sunnybrook Research Institute, Biological Sciences Platform, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Sisu Han
- Sunnybrook Research Institute, Biological Sciences Platform, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Carol Schuurmans
- Sunnybrook Research Institute, Biological Sciences Platform, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
43
|
Yamagishi S, Bando Y, Sato K. Involvement of Netrins and Their Receptors in Neuronal Migration in the Cerebral Cortex. Front Cell Dev Biol 2021; 8:590009. [PMID: 33520982 PMCID: PMC7843923 DOI: 10.3389/fcell.2020.590009] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 12/23/2020] [Indexed: 12/17/2022] Open
Abstract
In mammals, excitatory cortical neurons develop from the proliferative epithelium and progenitor cells in the ventricular zone and subventricular zone, and migrate radially to the cortical plate, whereas inhibitory GABAergic interneurons are born in the ganglionic eminence and migrate tangentially. The migration of newly born cortical neurons is tightly regulated by both extracellular and intracellular signaling to ensure proper positioning and projections. Non-cell-autonomous extracellular molecules, such as growth factors, axon guidance molecules, extracellular matrix, and other ligands, play a role in cortical migration, either by acting as attractants or repellents. In this article, we review the guidance molecules that act as cell-cell recognition molecules for the regulation of neuronal migration, with a focus on netrin family proteins, their receptors, and related molecules, including neogenin, repulsive guidance molecules (RGMs), Down syndrome cell adhesion molecule (DSCAM), fibronectin leucine-rich repeat transmembrane proteins (FLRTs), and draxin. Netrin proteins induce attractive and repulsive signals depending on their receptors. For example, binding of netrin-1 to deleted in colorectal cancer (DCC), possibly together with Unc5, repels migrating GABAergic neurons from the ventricular zone of the ganglionic eminence, whereas binding to α3β1 integrin promotes cortical interneuron migration. Human genetic disorders associated with these and related guidance molecules, such as congenital mirror movements, schizophrenia, and bipolar disorder, are also discussed.
Collapse
Affiliation(s)
- Satoru Yamagishi
- Department of Organ and Tissue Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yuki Bando
- Department of Organ and Tissue Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Kohji Sato
- Department of Organ and Tissue Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
44
|
Klein R, Pasterkamp RJ. Recent advances in inter-cellular interactions during neural circuit assembly. Curr Opin Neurobiol 2021; 69:25-32. [PMID: 33383489 DOI: 10.1016/j.conb.2020.12.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/02/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022]
Abstract
Neural circuit assembly is regulated by inter-cellular interactions involving secreted and surface-tethered guidance molecules. Here, we review recent progress in our understanding of their mechanisms-of-action and biological effects. We discuss mechanisms through which the secreted cue Netrin-1 regulates neuron migration and highlight novel roles for axon-derived secreted guidance cues. We cover recent structural work at atomic resolution that provides new insights into the activation mechanisms of axon guidance receptors and into protein complexes containing cell adhesion molecules, such as Teneurin (Ten), Latrophilin (Lphn) and FLRT. Ten-Ten homophilic, Ten-Lphn heterophilic, and Ten-Lphn-FLRT tripartite complexes seem to elicit distinct context-dependent cellular responses. Seemingly opposite responses can also be triggered by the Eph/ephrin signaling system. Here, recent work provides a simple mechanism for the decision of forming a new synapse versus rejection of the pre-synaptic partner. These studies identify novel regulatory mechanisms and biological functions that will apply generally in developing neural systems.
Collapse
Affiliation(s)
- Rüdiger Klein
- Department Molecules-Signaling-Development, Max Planck Institute of Neurobiology, Am Klopferspitz 18, 82152 Martinsried, Germany.
| | - Ronald Jeroen Pasterkamp
- Department of Translational Neuroscience, University Medical Center (UMC) Utrecht Brain Center, Utrecht University, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands.
| |
Collapse
|
45
|
Abstract
The mammalian cerebral cortex is the pinnacle of brain evolution, reaching its maximum complexity in terms of neuron number, diversity and functional circuitry. The emergence of this outstanding complexity begins during embryonic development, when a limited number of neural stem and progenitor cells manage to generate myriads of neurons in the appropriate numbers, types and proportions, in a process called neurogenesis. Here we review the current knowledge on the regulation of cortical neurogenesis, beginning with a description of the types of progenitor cells and their lineage relationships. This is followed by a review of the determinants of neuron fate, the molecular and genetic regulatory mechanisms, and considerations on the evolution of cortical neurogenesis in vertebrates leading to humans. We finish with an overview on how dysregulation of neurogenesis is a leading cause of human brain malformations and functional disabilities.
Collapse
Affiliation(s)
- Ana Villalba
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant, Spain
| | - Magdalena Götz
- Institute for Stem Cell Research, Helmholtz Zentrum München & Biomedical Center, Ludwig-Maximilians Universitaet, Planegg-Martinsried, Germany
| | - Víctor Borrell
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, Sant Joan d'Alacant, Spain.
| |
Collapse
|
46
|
AVŞAR T, ÇALIŞ Ş, YILMAZ B, DEMİRCİ OTLUOĞLU G, HOLYAVKİN C, KILIÇ T. Genome-wide identification of Chiari malformation type I associated candidate genes and chromosomal variations. Turk J Biol 2020; 44:449-456. [PMID: 33402871 PMCID: PMC7759189 DOI: 10.3906/biy-2009-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 10/12/2020] [Indexed: 01/12/2023] Open
Abstract
Chiari malformation type I (CMI) is a brain malformation that is characterized by herniation of the cerebellum into the spinal canal. Chiari malformation type I is highly heterogeneous; therefore, an accurate explanation of the pathogenesis of the disease is often not possible. Although some studies showed the role of genetics in CMI, the involvement of genetic variations in CMI pathogenesis has not been thoroughly elucidated. Therefore, in the current study we aim to reveal CMI-associated genomic variations in familial cases.Four CMI patients and 7 unaffected healthy members of two distinct families were analyzed. A microarray analysis of the affected and unaffected individuals from two Turkish families with CMI was conducted. Analyses of single nucleotide variations (SNVs) and copy number variations (CNVs) were performed by calculation of B allele frequency (BAF) and log R ratio (LRR) values from whole genome SNV data. Two missense variations, OLFML2A (rs7874348) and SLC4A9 (rs6860077), and a 5'UTR variation of COL4A1 (rs9521687) were significantly associated with CMI. Moreover, 12 SNVs in the intronic regions of FAM155A, NR3C1, TRPC7, ASTN2, and TRAF1 were determined to be associated with CMI. The CNV analysis showed that the 11p15.4 chromosome region is inherited in one of the families. The use of familial studies to explain the molecular pathogenesis of complex diseases such as CMI is crucial. It has been suggested that variations in OLFML2A, SLC4A9, and COL4A1 play a role in CMI molecular pathogenesis. The CNV analysis of individuals in both families revealed a potential chromosomal region, 11p15.4, and risk regions that are associated with CMI.
Collapse
Affiliation(s)
- Timuçin AVŞAR
- Department of Medical Biology, School of Medicine, Bahçeşehir University, İstanbulTurkey
- Neuroscience Program, Health Sciences Institute, Bahçeşehir University, İstanbulTurkey
- Neuroscience Laboratory, Health Sciences Institute, Bahçeşehir University, İstanbulTurkey
| | - Şeyma ÇALIŞ
- Neuroscience Laboratory, Health Sciences Institute, Bahçeşehir University, İstanbulTurkey
- Molecular Biology, Genetics, and Biotechnology Graduate Program, Graduate School of Science, Engineering, and Technology, İstanbul Technical University, İstanbulTurkey
| | - Baran YILMAZ
- Department of Neurosurgery, School of Medicine, Bahçeşehir University, İstanbulTurkey
| | | | - Can HOLYAVKİN
- Molecular Biology, Genetics, and Biotechnology Graduate Program, Graduate School of Science, Engineering, and Technology, İstanbul Technical University, İstanbulTurkey
| | - Türker KILIÇ
- Neuroscience Program, Health Sciences Institute, Bahçeşehir University, İstanbulTurkey
- Neuroscience Laboratory, Health Sciences Institute, Bahçeşehir University, İstanbulTurkey
- Department of Neurosurgery, School of Medicine, Bahçeşehir University, İstanbulTurkey
| |
Collapse
|
47
|
Takeda H, Kameo Y, Adachi T. Continuum modeling for neuronal lamination during cerebral morphogenesis considering cell migration and tissue growth. Comput Methods Biomech Biomed Engin 2020; 24:1-7. [PMID: 33290089 DOI: 10.1080/10255842.2020.1852554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/09/2020] [Accepted: 11/15/2020] [Indexed: 10/22/2022]
Abstract
For neuronal lamination during cerebral morphogenesis, later-born neurons must migrate through already-accumulated neurons. This neuronal migration is biochemically regulated by signaling molecules and mechanically affected by tissue deformation. To understand the neuronal lamination mechanisms, we constructed a continuum model of neuronal migration in a growing deformable tissue. We performed numerical analyses considering the migration promoted by signaling molecules and the tissue growth induced by neuron accumulation. The results suggest that the promoted migration and the space ensured by tissue growth are essential for neuronal lamination. The proposed model can describe the coupling of mechanical and biochemical mechanisms for neuronal lamination.
Collapse
Affiliation(s)
- Hironori Takeda
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto, Japan
- Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yoshitaka Kameo
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto, Japan
- Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
- Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Taiji Adachi
- Department of Micro Engineering, Graduate School of Engineering, Kyoto University, Kyoto, Japan
- Department of Biosystems Science, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
- Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| |
Collapse
|
48
|
Amin S, Borrell V. The Extracellular Matrix in the Evolution of Cortical Development and Folding. Front Cell Dev Biol 2020; 8:604448. [PMID: 33344456 PMCID: PMC7744631 DOI: 10.3389/fcell.2020.604448] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/12/2020] [Indexed: 02/02/2023] Open
Abstract
The evolution of the mammalian cerebral cortex leading to humans involved a remarkable sophistication of developmental mechanisms. Specific adaptations of progenitor cell proliferation and neuronal migration mechanisms have been proposed to play major roles in this evolution of neocortical development. One of the central elements influencing neocortex development is the extracellular matrix (ECM). The ECM provides both a structural framework during tissue formation and to present signaling molecules to cells, which directly influences cell behavior and movement. Here we review recent advances in the understanding of the role of ECM molecules on progenitor cell proliferation and neuronal migration, and how these contribute to cerebral cortex expansion and folding. We discuss how transcriptomic studies in human, ferret and mouse identify components of ECM as being candidate key players in cortex expansion during development and evolution. Then we focus on recent functional studies showing that ECM components regulate cortical progenitor cell proliferation, neuron migration and the mechanical properties of the developing cortex. Finally, we discuss how these features differ between lissencephalic and gyrencephalic species, and how the molecular evolution of ECM components and their expression profiles may have been fundamental in the emergence and evolution of cortex folding across mammalian phylogeny.
Collapse
Affiliation(s)
| | - Víctor Borrell
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Sant Joan d’Alacant, Spain
| |
Collapse
|
49
|
Abstract
The extraordinary diversity, variability, and complexity of cell types in the vertebrate brain is overwhelming and far exceeds that of any other organ. This complexity is the result of multiple cell divisions and intricate gene regulation and cell movements that take place during embryonic development. Understanding the cellular and molecular mechanisms underlying these complicated developmental processes requires the ability to obtain a complete registry of interconnected events often taking place far apart from each other. To assist with this challenging task, developmental neuroscientists take advantage of a broad set of methods and technologies, often adopted from other fields of research. Here, we review some of the methods developed in recent years whose use has rapidly spread for application in the field of developmental neuroscience. We also provide several considerations regarding the promise that these techniques hold for the near future and share some ideas on how existing methods from other research fields could help with the analysis of how neural circuits emerge.
Collapse
Affiliation(s)
- Augusto Escalante
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), Campus San Juan, Av. Ramón y Cajal s/n, Alicante 03550, Spain
| | - Rocío González-Martínez
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), Campus San Juan, Av. Ramón y Cajal s/n, Alicante 03550, Spain
| | - Eloísa Herrera
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), Campus San Juan, Av. Ramón y Cajal s/n, Alicante 03550, Spain
| |
Collapse
|
50
|
Lin Y, Yang J, Shen Z, Ma J, Simons BD, Shi SH. Behavior and lineage progression of neural progenitors in the mammalian cortex. Curr Opin Neurobiol 2020; 66:144-157. [PMID: 33227588 DOI: 10.1016/j.conb.2020.10.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/19/2020] [Accepted: 10/22/2020] [Indexed: 12/17/2022]
Abstract
The cerebral cortex is a central structure in the mammalian brain that enables higher cognitive functions and intellectual skills. It is the hallmark of the mammalian nervous system with enormous complexity, consisting of a large number of neurons and glia that are diverse in morphology, molecular expression, biophysical properties, circuit connectivity and physiological function. Cortical neurons and glia are generated by neural progenitor cells during development. Ensuring the correct cell cycle kinetics, fate behavior and lineage progression of neural progenitor cells is essential to determine the number and types of neurons and glia in the cerebral cortex, which together constitute neural circuits for brain function. In this review, we discuss recent findings on mammalian cortical progenitor cell types and their lineage behaviors in generating neurons and glia, cortical evolution and expansion, and advances in brain organoid technology that allow the modeling of human cortical development under normal and disease conditions.
Collapse
Affiliation(s)
- Yang Lin
- IDG/McGovern Institute for Brain Research, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center of Biological Structure, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jiajun Yang
- IDG/McGovern Institute for Brain Research, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center of Biological Structure, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zhongfu Shen
- IDG/McGovern Institute for Brain Research, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center of Biological Structure, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jian Ma
- IDG/McGovern Institute for Brain Research, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center of Biological Structure, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Benjamin D Simons
- The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK; Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, Wilberforce Road, Cambridge CB3 0WA, UK
| | - Song-Hai Shi
- IDG/McGovern Institute for Brain Research, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center of Biological Structure, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|