1
|
Lynch CJ, Elbau IG, Ng T, Ayaz A, Zhu S, Wolk D, Manfredi N, Johnson M, Chang M, Chou J, Summerville I, Ho C, Lueckel M, Bukhari H, Buchanan D, Victoria LW, Solomonov N, Goldwaser E, Moia S, Caballero-Gaudes C, Downar J, Vila-Rodriguez F, Daskalakis ZJ, Blumberger DM, Kay K, Aloysi A, Gordon EM, Bhati MT, Williams N, Power JD, Zebley B, Grosenick L, Gunning FM, Liston C. Frontostriatal salience network expansion in individuals in depression. Nature 2024; 633:624-633. [PMID: 39232159 PMCID: PMC11410656 DOI: 10.1038/s41586-024-07805-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 07/09/2024] [Indexed: 09/06/2024]
Abstract
Decades of neuroimaging studies have shown modest differences in brain structure and connectivity in depression, hindering mechanistic insights or the identification of risk factors for disease onset1. Furthermore, whereas depression is episodic, few longitudinal neuroimaging studies exist, limiting understanding of mechanisms that drive mood-state transitions. The emerging field of precision functional mapping has used densely sampled longitudinal neuroimaging data to show behaviourally meaningful differences in brain network topography and connectivity between and in healthy individuals2-4, but this approach has not been applied in depression. Here, using precision functional mapping and several samples of deeply sampled individuals, we found that the frontostriatal salience network is expanded nearly twofold in the cortex of most individuals with depression. This effect was replicable in several samples and caused primarily by network border shifts, with three distinct modes of encroachment occurring in different individuals. Salience network expansion was stable over time, unaffected by mood state and detectable in children before the onset of depression later in adolescence. Longitudinal analyses of individuals scanned up to 62 times over 1.5 years identified connectivity changes in frontostriatal circuits that tracked fluctuations in specific symptoms and predicted future anhedonia symptoms. Together, these findings identify a trait-like brain network topology that may confer risk for depression and mood-state-dependent connectivity changes in frontostriatal circuits that predict the emergence and remission of depressive symptoms over time.
Collapse
Affiliation(s)
- Charles J Lynch
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA.
| | - Immanuel G Elbau
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| | - Tommy Ng
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| | - Aliza Ayaz
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| | - Shasha Zhu
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| | - Danielle Wolk
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| | - Nicola Manfredi
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| | - Megan Johnson
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| | - Megan Chang
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| | - Jolin Chou
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| | | | - Claire Ho
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| | - Maximilian Lueckel
- Leibniz Institute for Resilience Research, Mainz, Germany
- Neuroimaging Center (NIC), Focus Program Translational Neurosciences (FTN), Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Hussain Bukhari
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| | - Derrick Buchanan
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | | | - Nili Solomonov
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| | - Eric Goldwaser
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| | - Stefano Moia
- Neuro-X Institute, École Polytechnique Fédérale de Lausanne, Geneva, Switzerland
- Department of Radiology and Medical Informatics, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Basque Center on Cognition, Brain and Language, Donostia, Spain
| | | | - Jonathan Downar
- Department of Psychiatry and Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Fidel Vila-Rodriguez
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Daniel M Blumberger
- Department of Psychiatry and Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Temerty Centre for Therapeutic Brain Intervention, Toronto, Ontario, Canada
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Kendrick Kay
- Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA
| | - Amy Aloysi
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Evan M Gordon
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Mahendra T Bhati
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Nolan Williams
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Jonathan D Power
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| | - Benjamin Zebley
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| | - Logan Grosenick
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| | - Faith M Gunning
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| | - Conor Liston
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
2
|
Chowdhury G, Umeda K, Ohyanagi T, Nasu K, Yamasu K. Involvement of nr2f genes in brain regionalization and eye development during early zebrafish development. Dev Growth Differ 2024; 66:145-160. [PMID: 38263801 PMCID: PMC11457503 DOI: 10.1111/dgd.12912] [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: 11/27/2023] [Revised: 01/02/2024] [Accepted: 01/06/2024] [Indexed: 01/25/2024]
Abstract
Nuclear receptor subfamily 2 group F (Nr2f) proteins are essential for brain development in mice, but little is known about their precise roles and their evolutionary diversification. In the present study, the expression patterns of major nr2f genes (nr2f1a, nr2f1b, and nr2f2) during early brain development were investigated in zebrafish. Comparisons of their expression patterns revealed similar but temporally and spatially distinct patterns after early somite stages in the brain. Frameshift mutations in the three nr2f genes, achieved using the CRISPR/Cas9 method, resulted in a smaller telencephalon and smaller eyes in the nr2f1a mutants; milder forms of those defects were present in the nr2f1b and nr2f2 mutants. Acridine orange staining revealed enhanced cell death in the brain and/or eyes in all nr2f homozygous mutants. The expression of regional markers in the brain did not suggest global defects in brain regionalization; however, shha expression in the preoptic area and hypothalamus, as well as fgf8a expression in the anterior telencephalon, was disturbed in nr2f1a and nr2f1b mutants, potentially leading to a defective telencephalon. Specification of the retina and optic stalk was also significantly affected. The overexpression of nr2f1b by injection of mRNA disrupted the anterior brain at a high dose, and the expression of pax6a in the eyes and fgf8a in the telencephalon at a low dose. The results of these loss- and gain-of-function approaches showed that nr2f genes regulate the development of the telencephalon and eyes in zebrafish embryos.
Collapse
Affiliation(s)
- Gazlima Chowdhury
- Division of Life Science, Graduate School of Science and EngineeringSaitama UniversitySaitamaJapan
- Department of Aquatic Environment and Resource ManagementSher‐e‐Bangla Agricultural UniversityDhakaBangladesh
| | - Koto Umeda
- Division of Life Science, Graduate School of Science and EngineeringSaitama UniversitySaitamaJapan
| | - Takero Ohyanagi
- Division of Life Science, Graduate School of Science and EngineeringSaitama UniversitySaitamaJapan
| | - Kouhei Nasu
- Division of Life Science, Graduate School of Science and EngineeringSaitama UniversitySaitamaJapan
| | - Kyo Yamasu
- Division of Life Science, Graduate School of Science and EngineeringSaitama UniversitySaitamaJapan
| |
Collapse
|
3
|
Molnár Z, Kwan KY. Development and Evolution of Thalamocortical Connectivity. Cold Spring Harb Perspect Biol 2024; 16:a041503. [PMID: 38167425 PMCID: PMC10759993 DOI: 10.1101/cshperspect.a041503] [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] [Indexed: 01/05/2024]
Abstract
Conscious perception in mammals depends on precise circuit connectivity between cerebral cortex and thalamus; the evolution and development of these structures are closely linked. During the wiring of reciprocal thalamus-cortex connections, thalamocortical axons (TCAs) first navigate forebrain regions that had undergone substantial evolutionary modifications. In particular, the organization of the pallial-subpallial boundary (PSPB) diverged significantly between mammals, reptiles, and birds. In mammals, transient cell populations in internal capsule and early corticofugal projections from subplate neurons closely interact with TCAs to guide pathfinding through ventral forebrain and PSPB crossing. Prior to thalamocortical axon arrival, cortical areas are initially patterned by intrinsic genetic factors. Thalamocortical axons then innervate cortex in a topographically organized manner to enable sensory input to refine cortical arealization. Here, we review the mechanisms underlying the guidance of thalamocortical axons across forebrain boundaries, the implications of PSPB evolution for thalamocortical axon pathfinding, and the reciprocal influence between thalamus and cortex during development.
Collapse
Affiliation(s)
- Zoltán Molnár
- Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Oxford OX1 3PT, United Kingdom
| | - Kenneth Y Kwan
- Michigan Neuroscience Institute (MNI), Department of Human Genetics, University of Michigan, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
4
|
Zheng Y, Zhao C, Song Q, Xu L, Zhang B, Hu G, Kong X, Li S, Li X, Shen Y, Zhuang L, Wu M, Liu Y, Zhou Y. Histone methylation mediated by NSD1 is required for the establishment and maintenance of neuronal identities. Cell Rep 2023; 42:113496. [PMID: 37995181 DOI: 10.1016/j.celrep.2023.113496] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 09/28/2023] [Accepted: 11/09/2023] [Indexed: 11/25/2023] Open
Abstract
Appropriate histone modifications emerge as essential cell fate regulators of neuronal identities across neocortical areas and layers. Here we showed that NSD1, the methyltransferase for di-methylated lysine 36 of histone H3 (H3K36me2), controls both area and layer identities of the neocortex. Nsd1-ablated neocortex showed an area shift of all four primary functional regions and aberrant wiring of cortico-thalamic-cortical projections. Nsd1 conditional knockout mice displayed defects in spatial memory, motor learning, and coordination, resembling patients with the Sotos syndrome carrying NSD1 mutations. On Nsd1 loss, superficial-layer pyramidal neurons (PNs) progressively mis-expressed markers for deep-layer PNs, and PNs remained immature both morphologically and electrophysiologically. Loss of Nsd1 in postmitotic PNs causes genome-wide loss of H3K36me2 and re-distribution of DNA methylation, which accounts for diminished expression of neocortical layer specifiers but ectopic expression of non-neural genes. Together, H3K36me2 mediated by NSD1 is required for the establishment and maintenance of region- and layer-specific neocortical identities.
Collapse
Affiliation(s)
- Yue Zheng
- Department of Neurosurgery, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan 430071, China
| | - Chen Zhao
- Department of Neurosurgery, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan 430071, China
| | - Qiulin Song
- Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan 430071, China; Eye Center, Wuhan University Renmin Hospital, Wuhan 430071, China
| | - Lichao Xu
- Department of Neurosurgery, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan 430071, China
| | - Bo Zhang
- Department of Neurosurgery, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan 430071, China
| | - Guangda Hu
- Department of Neurosurgery, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan 430071, China
| | - Xiangfei Kong
- Department of Neurosurgery, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan 430071, China
| | - Shaowen Li
- Department of Neurosurgery, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan 430071, China
| | - Xiang Li
- Department of Neurosurgery, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan 430071, China
| | - Yin Shen
- Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan 430071, China; Eye Center, Wuhan University Renmin Hospital, Wuhan 430071, China
| | - Lenan Zhuang
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Min Wu
- Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan 430071, China; College of Life Sciences, Taikang Center for Life and Medical Sciences of Wuhan University, Wuhan 430071, China.
| | - Ying Liu
- Department of Neurosurgery, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan 430071, China.
| | - Yan Zhou
- Department of Neurosurgery, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; Frontier Science Center of Immunology and Metabolism, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
5
|
Jorstad NL, Close J, Johansen N, Yanny AM, Barkan ER, Travaglini KJ, Bertagnolli D, Campos J, Casper T, Crichton K, Dee N, Ding SL, Gelfand E, Goldy J, Hirschstein D, Kiick K, Kroll M, Kunst M, Lathia K, Long B, Martin N, McMillen D, Pham T, Rimorin C, Ruiz A, Shapovalova N, Shehata S, Siletti K, Somasundaram S, Sulc J, Tieu M, Torkelson A, Tung H, Callaway EM, Hof PR, Keene CD, Levi BP, Linnarsson S, Mitra PP, Smith K, Hodge RD, Bakken TE, Lein ES. Transcriptomic cytoarchitecture reveals principles of human neocortex organization. Science 2023; 382:eadf6812. [PMID: 37824655 DOI: 10.1126/science.adf6812] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 09/08/2023] [Indexed: 10/14/2023]
Abstract
Variation in cytoarchitecture is the basis for the histological definition of cortical areas. We used single cell transcriptomics and performed cellular characterization of the human cortex to better understand cortical areal specialization. Single-nucleus RNA-sequencing of 8 areas spanning cortical structural variation showed a highly consistent cellular makeup for 24 cell subclasses. However, proportions of excitatory neuron subclasses varied substantially, likely reflecting differences in connectivity across primary sensorimotor and association cortices. Laminar organization of astrocytes and oligodendrocytes also differed across areas. Primary visual cortex showed characteristic organization with major changes in the excitatory to inhibitory neuron ratio, expansion of layer 4 excitatory neurons, and specialized inhibitory neurons. These results lay the groundwork for a refined cellular and molecular characterization of human cortical cytoarchitecture and areal specialization.
Collapse
Affiliation(s)
| | - Jennie Close
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | - Eliza R Barkan
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | - Jazmin Campos
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Tamara Casper
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Song-Lin Ding
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Emily Gelfand
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Katelyn Kiick
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Matthew Kroll
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Michael Kunst
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Kanan Lathia
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Brian Long
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Naomi Martin
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | | | - Augustin Ruiz
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Soraya Shehata
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Kimberly Siletti
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | - Josef Sulc
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Michael Tieu
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Amy Torkelson
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Herman Tung
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Edward M Callaway
- Systems Neurobiology Laboratories, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Patrick R Hof
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - C Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Boaz P Levi
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | - Sten Linnarsson
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Partha P Mitra
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY 11724, USA
| | - Kimberly Smith
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | | | - Ed S Lein
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| |
Collapse
|
6
|
Lynch CJ, Elbau I, Ng T, Ayaz A, Zhu S, Manfredi N, Johnson M, Wolk D, Power JD, Gordon EM, Kay K, Aloysi A, Moia S, Caballero-Gaudes C, Victoria LW, Solomonov N, Goldwaser E, Zebley B, Grosenick L, Downar J, Vila-Rodriguez F, Daskalakis ZJ, Blumberger DM, Williams N, Gunning FM, Liston C. Expansion of a frontostriatal salience network in individuals with depression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.09.551651. [PMID: 37645792 PMCID: PMC10461904 DOI: 10.1101/2023.08.09.551651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Hundreds of neuroimaging studies spanning two decades have revealed differences in brain structure and functional connectivity in depression, but with modest effect sizes, complicating efforts to derive mechanistic pathophysiologic insights or develop biomarkers. 1 Furthermore, although depression is a fundamentally episodic condition, few neuroimaging studies have taken a longitudinal approach, which is critical for understanding cause and effect and delineating mechanisms that drive mood state transitions over time. The emerging field of precision functional mapping using densely-sampled longitudinal neuroimaging data has revealed unexpected, functionally meaningful individual differences in brain network topology in healthy individuals, 2-5 but these approaches have never been applied to individuals with depression. Here, using precision functional mapping techniques and 11 datasets comprising n=187 repeatedly sampled individuals and >21,000 minutes of fMRI data, we show that the frontostriatal salience network is expanded two-fold in most individuals with depression. This effect was replicable in multiple samples, including large-scale, group-average data (N=1,231 subjects), and caused primarily by network border shifts affecting specific functional systems, with three distinct modes of encroachment occurring in different individuals. Salience network expansion was unexpectedly stable over time, unaffected by changes in mood state, and detectable in children before the subsequent onset of depressive symptoms in adolescence. Longitudinal analyses of individuals scanned up to 62 times over 1.5 years identified connectivity changes in specific frontostriatal circuits that tracked fluctuations in specific symptom domains and predicted future anhedonia symptoms before they emerged. Together, these findings identify a stable trait-like brain network topology that may confer risk for depression and mood-state dependent connectivity changes in frontostriatal circuits that predict the emergence and remission of depressive symptoms over time.
Collapse
|
7
|
de Sousa AA, Beaudet A, Calvey T, Bardo A, Benoit J, Charvet CJ, Dehay C, Gómez-Robles A, Gunz P, Heuer K, van den Heuvel MP, Hurst S, Lauters P, Reed D, Salagnon M, Sherwood CC, Ströckens F, Tawane M, Todorov OS, Toro R, Wei Y. From fossils to mind. Commun Biol 2023; 6:636. [PMID: 37311857 PMCID: PMC10262152 DOI: 10.1038/s42003-023-04803-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 04/04/2023] [Indexed: 06/15/2023] Open
Abstract
Fossil endocasts record features of brains from the past: size, shape, vasculature, and gyrification. These data, alongside experimental and comparative evidence, are needed to resolve questions about brain energetics, cognitive specializations, and developmental plasticity. Through the application of interdisciplinary techniques to the fossil record, paleoneurology has been leading major innovations. Neuroimaging is shedding light on fossil brain organization and behaviors. Inferences about the development and physiology of the brains of extinct species can be experimentally investigated through brain organoids and transgenic models based on ancient DNA. Phylogenetic comparative methods integrate data across species and associate genotypes to phenotypes, and brains to behaviors. Meanwhile, fossil and archeological discoveries continuously contribute new knowledge. Through cooperation, the scientific community can accelerate knowledge acquisition. Sharing digitized museum collections improves the availability of rare fossils and artifacts. Comparative neuroanatomical data are available through online databases, along with tools for their measurement and analysis. In the context of these advances, the paleoneurological record provides ample opportunity for future research. Biomedical and ecological sciences can benefit from paleoneurology's approach to understanding the mind as well as its novel research pipelines that establish connections between neuroanatomy, genes and behavior.
Collapse
Affiliation(s)
| | - Amélie Beaudet
- Laboratoire de Paléontologie, Évolution, Paléoécosystèmes et Paléoprimatologie (PALEVOPRIM), UMR 7262 CNRS & Université de Poitiers, Poitiers, France.
- University of Cambridge, Cambridge, UK.
| | - Tanya Calvey
- Division of Clinical Anatomy and Biological Anthropology, University of Cape Town, Cape Town, South Africa.
| | - Ameline Bardo
- UMR 7194, CNRS-MNHN, Département Homme et Environnement, Musée de l'Homme, Paris, France
- Skeletal Biology Research Centre, School of Anthropology and Conservation, University of Kent, Canterbury, UK
| | - Julien Benoit
- Evolutionary Studies Institute, University of the Witwatersrand, Johannesburg, South Africa
| | - Christine J Charvet
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, USA
| | - Colette Dehay
- University of Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, F-69500, Bron, France
| | | | - Philipp Gunz
- Department of Human Origins, Max Planck Institute for Evolutionary Anthropology, Deutscher Platz 6, D-04103, Leipzig, Germany
| | - Katja Heuer
- Institut Pasteur, Université Paris Cité, Unité de Neuroanatomie Appliquée et Théorique, F-75015, Paris, France
| | | | - Shawn Hurst
- University of Indianapolis, Indianapolis, IN, USA
| | - Pascaline Lauters
- Institut royal des Sciences naturelles, Direction Opérationnelle Terre et Histoire de la Vie, Brussels, Belgium
| | - Denné Reed
- Department of Anthropology, University of Texas at Austin, Austin, TX, USA
| | - Mathilde Salagnon
- CNRS, CEA, IMN, GIN, UMR 5293, Université Bordeaux, Bordeaux, France
- PACEA UMR 5199, CNRS, Université Bordeaux, Pessac, France
| | - Chet C Sherwood
- Department of Anthropology, The George Washington University, Washington, DC, USA
| | - Felix Ströckens
- C. & O. Vogt Institute for Brain Research, University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Mirriam Tawane
- Ditsong National Museum of Natural History, Pretoria, South Africa
| | - Orlin S Todorov
- School of Natural Sciences, Macquarie University, Sydney, NSW, Australia
| | - Roberto Toro
- Institut Pasteur, Université Paris Cité, Unité de Neuroanatomie Appliquée et Théorique, F-75015, Paris, France
| | - Yongbin Wei
- Beijing University of Posts and Telecommunications, Beijing, China
| |
Collapse
|
8
|
Sen SQ. Generating neural diversity through spatial and temporal patterning. Semin Cell Dev Biol 2023; 142:54-66. [PMID: 35738966 DOI: 10.1016/j.semcdb.2022.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 06/08/2022] [Accepted: 06/08/2022] [Indexed: 11/19/2022]
Abstract
The nervous system consists of a vast diversity of neurons and glia that are accurately assembled into functional circuits. What are the mechanisms that generate these diverse cell types? During development, an epithelial sheet with neurogenic potential is initially regionalised into spatially restricted domains of gene expression. From this, pools of neural stem cells (NSCs) with distinct molecular profiles and the potential to generate different neuron types, are specified. These NSCs then divide asymmetrically to self-renew and generate post-mitotic neurons or glia. As NSCs age, they experience transitions in gene expression, which further allows them to generate different neurons or glia over time. Versions of this general template of spatial and temporal patterning operate during the development of different parts of different nervous systems. Here, I cover our current knowledge of Drosophila brain and optic lobe development as well as the development of the vertebrate cortex and spinal cord within the framework of this above template. I highlight where our knowledge is lacking, where mechanisms beyond these might operate, and how the emergence of new technologies might help address unanswered questions.
Collapse
Affiliation(s)
- Sonia Q Sen
- Tata Institute for Genetics and Society, UAS-GKVK Campus, Bellary Road, Bangalore, India.
| |
Collapse
|
9
|
Samara A, Spildrejorde M, Sharma A, Falck M, Leithaug M, Modafferi S, Bjørnstad PM, Acharya G, Gervin K, Lyle R, Eskeland R. A multi-omics approach to visualize early neuronal differentiation from hESCs in 4D. iScience 2022; 25:105279. [PMID: 36304110 PMCID: PMC9593815 DOI: 10.1016/j.isci.2022.105279] [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: 03/01/2022] [Revised: 08/22/2022] [Accepted: 09/28/2022] [Indexed: 11/19/2022] Open
Abstract
Neuronal differentiation of pluripotent stem cells is an established method to study physiology, disease, and medication safety. However, the sequence of events in human neuronal differentiation and the ability of in vitro models to recapitulate early brain development are poorly understood. We developed a protocol optimized for the study of early human brain development and neuropharmacological applications. We comprehensively characterized gene expression and epigenetic profiles at four timepoints, because the cells differentiate from embryonic stem cells towards a heterogeneous population of progenitors, immature and mature neurons bearing telencephalic signatures. A multi-omics roadmap of neuronal differentiation, combined with searchable interactive gene analysis tools, allows for extensive exploration of early neuronal development and the effect of medications.
Collapse
Affiliation(s)
- Athina Samara
- Division of Clinical Paediatrics, Department of Women’s and Children’s Health, Karolinska Institutet, Solna, Sweden
- Astrid Lindgren Children′s Hospital Karolinska University Hospital, Stockholm, Sweden
| | - Mari Spildrejorde
- PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Ankush Sharma
- Department of Informatics, University of Oslo, Oslo, Norway
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Martin Falck
- PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Magnus Leithaug
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Stefania Modafferi
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Pål Marius Bjørnstad
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Ganesh Acharya
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Alfred Nobels Allé 8, SE-14152 Stockholm, Sweden
- Center for Fetal Medicine, Karolinska University Hospital Huddinge, SE-14186 Stockholm, Sweden
| | - Kristina Gervin
- PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
- Pharmacoepidemiology and Drug Safety Research Group, Department of Pharmacy, School of Pharmacy, University of Oslo, Oslo, Norway
- Division of Clinical Neuroscience, Department of Research and Innovation, Oslo University Hospital, Oslo, Norway
| | - Robert Lyle
- PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Ragnhild Eskeland
- PharmaTox Strategic Research Initiative, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
10
|
Nguyen H, Sokpor G, Parichha A, Pham L, Saikhedkar N, Xie Y, Ulmke PA, Rosenbusch J, Pirouz M, Behr R, Stoykova A, Brand-Saberi B, Nguyen HP, Staiger JF, Tole S, Tuoc T. BAF (mSWI/SNF) complex regulates mediolateral cortical patterning in the developing forebrain. Front Cell Dev Biol 2022; 10:1011109. [PMID: 36263009 PMCID: PMC9573979 DOI: 10.3389/fcell.2022.1011109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/16/2022] [Indexed: 11/24/2022] Open
Abstract
Early forebrain patterning entails the correct regional designation of the neuroepithelium, and appropriate specification, generation, and distribution of neural cells during brain development. Specific signaling and transcription factors are known to tightly regulate patterning of the dorsal telencephalon to afford proper structural/functional cortical arealization and morphogenesis. Nevertheless, whether and how changes of the chromatin structure link to the transcriptional program(s) that control cortical patterning remains elusive. Here, we report that the BAF chromatin remodeling complex regulates the spatiotemporal patterning of the mouse dorsal telencephalon. To determine whether and how the BAF complex regulates cortical patterning, we conditionally deleted the BAF complex scaffolding subunits BAF155 and BAF170 in the mouse dorsal telencephalic neuroepithelium. Morphological and cellular changes in the BAF mutant forebrain were examined using immunohistochemistry and in situ hybridization. RNA sequencing, Co-immunoprecipitation, and mass spectrometry were used to investigate the molecular basis of BAF complex involvement in forebrain patterning. We found that conditional ablation of BAF complex in the dorsal telencephalon neuroepithelium caused expansion of the cortical hem and medial cortex beyond their developmental boundaries. Consequently, the hippocampal primordium is not specified, the mediolateral cortical patterning is compromised, and the cortical identity is disturbed in the absence of BAF complex. The BAF complex was found to interact with the cortical hem suppressor LHX2. The BAF complex suppresses cortical hem fate to permit proper forebrain patterning. We provide evidence that BAF complex modulates mediolateral cortical patterning possibly by interacting with the transcription factor LHX2 to drive the LHX2-dependent transcriptional program essential for dorsal telencephalon patterning. Our data suggest a putative mechanistic synergy between BAF chromatin remodeling complex and LHX2 in regulating forebrain patterning and ontogeny.
Collapse
Affiliation(s)
- Huong Nguyen
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
- Faculty of Biotechnology, Thai Nguyen University of Sciences, Thai Nguyen, Vietnam
| | - Godwin Sokpor
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, Bochum, Germany
| | | | - Linh Pham
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
| | | | - Yuanbin Xie
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
| | - Pauline Antonie Ulmke
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
| | - Joachim Rosenbusch
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
| | - Mehdi Pirouz
- Max Planck Institute for Multidisciplinary Sciences, Goettingen, Germany
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, United States
| | - Rüdiger Behr
- German Primate Center-Leibniz Institute for Primate Research, Goettingen, Germany
| | | | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, Bochum, Germany
| | - Huu Phuc Nguyen
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
| | - Jochen F. Staiger
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
| | - Shubha Tole
- Tata Institute of Fundamental Research, Mumbai, India
- *Correspondence: Shubha Tole, ; Tran Tuoc,
| | - Tran Tuoc
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Goettingen, Goettingen, Germany
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
- *Correspondence: Shubha Tole, ; Tran Tuoc,
| |
Collapse
|
11
|
Abstract
Immunity could be viewed as the common factor in neurodevelopmental disorders and cancer. The immune and nervous systems coevolve as the embryo develops. Immunity can release cytokines that activate MAPK signaling in neural cells. In specific embryonic brain cell types, dysregulated signaling that results from germline or embryonic mutations can promote changes in chromatin organization and gene accessibility, and thus expression levels of essential genes in neurodevelopment. In cancer, dysregulated signaling can emerge from sporadic somatic mutations during human life. Neurodevelopmental disorders and cancer share similarities. In neurodevelopmental disorders, immunity, and cancer, there appears an almost invariable involvement of small GTPases (e.g., Ras, RhoA, and Rac) and their pathways. TLRs, IL-1, GIT1, and FGFR signaling pathways, all can be dysregulated in neurodevelopmental disorders and cancer. Although there are signaling similarities, decisive differentiating factors are timing windows, and cell type specific perturbation levels, pointing to chromatin reorganization. Finally, we discuss drug discovery.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, USA
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
- Corresponding author
| | - Chung-Jung Tsai
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| |
Collapse
|
12
|
Sato H, Hatakeyama J, Iwasato T, Araki K, Yamamoto N, Shimamura K. Thalamocortical axons control the cytoarchitecture of neocortical layers by area-specific supply of VGF. eLife 2022; 11:67549. [PMID: 35289744 PMCID: PMC8959604 DOI: 10.7554/elife.67549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/12/2022] [Indexed: 11/13/2022] Open
Abstract
Neuronal abundance and thickness of each cortical layer are specific to each area, but how this fundamental feature arises during development remains poorly understood. While some of area-specific features are controlled by intrinsic cues such as morphogens and transcription factors, the exact influence and mechanisms of action by cues extrinsic to the cortex, in particular the thalamic axons, have not been fully established. Here, we identify a thalamus-derived factor, VGF, which is indispensable for thalamocortical axons to maintain the proper amount of layer 4 neurons in the mouse sensory cortices. This process is prerequisite for further maturation of the primary somatosensory area, such as barrel field formation instructed by a neuronal activity-dependent mechanism. Our results provide an actual case in which highly site-specific axon projection confers further regional complexity upon the target field through locally secreting signaling molecules from axon terminals.
Collapse
Affiliation(s)
- Haruka Sato
- Department of Brain Morphogenesis, Kumamoto University, Kumamoto, Japan
| | - Jun Hatakeyama
- Department of Brain Morphogenesis, Kumamoto University, Kumamoto, Japan
| | - Takuji Iwasato
- Laboratory of Mammalian Neural Circuits, National Institute of Genetics, Mishima, Japan
| | - Kimi Araki
- Department of Brain Morphogenesis, Kumamoto University, Kumamoto, Japan
| | - Nobuhiko Yamamoto
- Laboratory of Cellular and Molecular Neurobiology, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Kenji Shimamura
- Department of Brain Morphogenesis, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
13
|
Harb K, Richter M, Neelagandan N, Magrinelli E, Harfoush H, Kuechler K, Henis M, Hermanns-Borgmeyer I, Calderon de Anda F, Duncan K. Pum2 and TDP-43 refine area-specific cytoarchitecture post-mitotically and modulate translation of Sox5, Bcl11b, and Rorb mRNAs in developing mouse neocortex. eLife 2022; 11:55199. [PMID: 35262486 PMCID: PMC8906809 DOI: 10.7554/elife.55199] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 02/25/2022] [Indexed: 12/15/2022] Open
Abstract
In the neocortex, functionally distinct areas process specific types of information. Area identity is established by morphogens and transcriptional master regulators, but downstream mechanisms driving area-specific neuronal specification remain unclear. Here, we reveal a role for RNA-binding proteins in defining area-specific cytoarchitecture. Mice lacking Pum2 or overexpressing human TDP-43 show apparent ‘motorization’ of layers IV and V of primary somatosensory cortex (S1), characterized by dramatic expansion of cells co-expressing Sox5 and Bcl11b/Ctip2, a hallmark of subcerebral projection neurons, at the expense of cells expressing the layer IV neuronal marker Rorβ. Moreover, retrograde labeling experiments with cholera toxin B in Pum2; Emx1-Cre and TDP43A315T mice revealed a corresponding increase in subcerebral connectivity of these neurons in S1. Intriguingly, other key features of somatosensory area identity are largely preserved, suggesting that Pum2 and TDP-43 may function in a downstream program, rather than controlling area identity per se. Transfection of primary neurons and in utero electroporation (IUE) suggest cell-autonomous and post-mitotic modulation of Sox5, Bcl11b/Ctip2, and Rorβ levels. Mechanistically, we find that Pum2 and TDP-43 directly interact with and affect the translation of mRNAs encoding Sox5, Bcl11b/Ctip2, and Rorβ. In contrast, effects on the levels of these mRNAs were not detectable in qRT-PCR or single-molecule fluorescent in situ hybridization assays, and we also did not detect effects on their splicing or polyadenylation patterns. Our results support the notion that post-transcriptional regulatory programs involving translational regulation and mediated by Pum2 and TDP-43 contribute to elaboration of area-specific neuronal identity and connectivity in the neocortex.
Collapse
Affiliation(s)
- Kawssar Harb
- Neuronal Translational Control Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Melanie Richter
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nagammal Neelagandan
- Neuronal Translational Control Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Elia Magrinelli
- Department of Basic Neuroscience, University of Geneva, Geneva, Switzerland
| | - Hend Harfoush
- Neuronal Translational Control Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Katrin Kuechler
- Neuronal Translational Control Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Melad Henis
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Anatomy and Histology, Faculty of Veterinary Medicine, New Valley University, New Valley, Egypt
| | - Irm Hermanns-Borgmeyer
- Transgenic Service Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Froylan Calderon de Anda
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kent Duncan
- Neuronal Translational Control Group, Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| |
Collapse
|
14
|
Tocco C, Øvsthus M, Bjaalie JG, Leergaard TB, Studer M. The topography of corticopontine projections is controlled by postmitotic expression of the area-mapping gene Nr2f1. Development 2022; 149:274658. [PMID: 35262177 PMCID: PMC8959144 DOI: 10.1242/dev.200026] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 12/20/2021] [Indexed: 12/27/2022]
Abstract
Axonal projections from layer V neurons of distinct neocortical areas are topographically organized into discrete clusters within the pontine nuclei during the establishment of voluntary movements. However, the molecular determinants controlling corticopontine connectivity are insufficiently understood. Here, we show that an intrinsic cortical genetic program driven by Nr2f1 graded expression is directly implicated in the organization of corticopontine topographic mapping. Transgenic mice lacking cortical expression of Nr2f1 and exhibiting areal organization defects were used as model systems to investigate the arrangement of corticopontine projections. By combining three-dimensional digital brain atlas tools, Cre-dependent mouse lines and axonal tracing, we show that Nr2f1 expression in postmitotic neurons spatially and temporally controls somatosensory topographic projections, whereas expression in progenitor cells influences the ratio between corticopontine and corticospinal fibres passing the pontine nuclei. We conclude that cortical gradients of area-patterning genes are directly implicated in the establishment of a topographic somatotopic mapping from the cortex onto pontine nuclei. Summary: Cortical gradient expression of the area patterning gene Nr2f1 spatially and temporally controls corticopontine topographic connectivity in layer V projection neurons.
Collapse
Affiliation(s)
- Chiara Tocco
- University Côte d'Azur, CNRS, Inserm, iBV, Nice 06108, France
| | - Martin Øvsthus
- Institute of Basic Medical Sciences, University of Oslo, Oslo N-0317, Norway
| | - Jan G Bjaalie
- Institute of Basic Medical Sciences, University of Oslo, Oslo N-0317, Norway
| | - Trygve B Leergaard
- Institute of Basic Medical Sciences, University of Oslo, Oslo N-0317, Norway
| | - Michèle Studer
- University Côte d'Azur, CNRS, Inserm, iBV, Nice 06108, France
| |
Collapse
|
15
|
Tocco C, Bertacchi M, Studer M. Structural and Functional Aspects of the Neurodevelopmental Gene NR2F1: From Animal Models to Human Pathology. Front Mol Neurosci 2022; 14:767965. [PMID: 34975398 PMCID: PMC8715095 DOI: 10.3389/fnmol.2021.767965] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/25/2021] [Indexed: 01/28/2023] Open
Abstract
The assembly and maturation of the mammalian brain result from an intricate cascade of highly coordinated developmental events, such as cell proliferation, migration, and differentiation. Any impairment of this delicate multi-factorial process can lead to complex neurodevelopmental diseases, sharing common pathogenic mechanisms and molecular pathways resulting in multiple clinical signs. A recently described monogenic neurodevelopmental syndrome named Bosch-Boonstra-Schaaf Optic Atrophy Syndrome (BBSOAS) is caused by NR2F1 haploinsufficiency. The NR2F1 gene, coding for a transcriptional regulator belonging to the steroid/thyroid hormone receptor superfamily, is known to play key roles in several brain developmental processes, from proliferation and differentiation of neural progenitors to migration and identity acquisition of neocortical neurons. In a clinical context, the disruption of these cellular processes could underlie the pathogenesis of several symptoms affecting BBSOAS patients, such as intellectual disability, visual impairment, epilepsy, and autistic traits. In this review, we will introduce NR2F1 protein structure, molecular functioning, and expression profile in the developing mouse brain. Then, we will focus on Nr2f1 several functions during cortical development, from neocortical area and cell-type specification to maturation of network activity, hippocampal development governing learning behaviors, assembly of the visual system, and finally establishment of cortico-spinal descending tracts regulating motor execution. Whenever possible, we will link experimental findings in animal or cellular models to corresponding features of the human pathology. Finally, we will highlight some of the unresolved questions on the diverse functions played by Nr2f1 during brain development, in order to propose future research directions. All in all, we believe that understanding BBSOAS mechanisms will contribute to further unveiling pathophysiological mechanisms shared by several neurodevelopmental disorders and eventually lead to effective treatments.
Collapse
Affiliation(s)
- Chiara Tocco
- Université Côte d'Azur, CNRS, Inserm, iBV, Nice, France
| | | | | |
Collapse
|
16
|
Shibata M, Pattabiraman K, Lorente-Galdos B, Andrijevic D, Kim SK, Kaur N, Muchnik SK, Xing X, Santpere G, Sousa AMM, Sestan N. Regulation of prefrontal patterning and connectivity by retinoic acid. Nature 2021; 598:483-488. [PMID: 34599305 PMCID: PMC9018119 DOI: 10.1038/s41586-021-03953-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 08/25/2021] [Indexed: 02/08/2023]
Abstract
The prefrontal cortex (PFC) and its connections with the mediodorsal thalamus are crucial for cognitive flexibility and working memory1 and are thought to be altered in disorders such as autism2,3 and schizophrenia4,5. Although developmental mechanisms that govern the regional patterning of the cerebral cortex have been characterized in rodents6-9, the mechanisms that underlie the development of PFC-mediodorsal thalamus connectivity and the lateral expansion of the PFC with a distinct granular layer 4 in primates10,11 remain unknown. Here we report an anterior (frontal) to posterior (temporal), PFC-enriched gradient of retinoic acid, a signalling molecule that regulates neural development and function12-15, and we identify genes that are regulated by retinoic acid in the neocortex of humans and macaques at the early and middle stages of fetal development. We observed several potential sources of retinoic acid, including the expression and cortical expansion of retinoic-acid-synthesizing enzymes specifically in primates as compared to mice. Furthermore, retinoic acid signalling is largely confined to the prospective PFC by CYP26B1, a retinoic-acid-catabolizing enzyme, which is upregulated in the prospective motor cortex. Genetic deletions in mice revealed that retinoic acid signalling through the retinoic acid receptors RXRG and RARB, as well as CYP26B1-dependent catabolism, are involved in proper molecular patterning of prefrontal and motor areas, development of PFC-mediodorsal thalamus connectivity, intra-PFC dendritic spinogenesis and expression of the layer 4 marker RORB. Together, these findings show that retinoic acid signalling has a critical role in the development of the PFC and, potentially, in its evolutionary expansion.
Collapse
Affiliation(s)
- Mikihito Shibata
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Kartik Pattabiraman
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Yale Child Study Center, Yale School of Medicine, New Haven, CT, USA
| | | | - David Andrijevic
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Suel-Kee Kim
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Navjot Kaur
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Sydney K Muchnik
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Xiaojun Xing
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Yale Genome Editing Center, Yale School of Medicine, New Haven, CT, USA
| | - Gabriel Santpere
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Neurogenomics Group, Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), DCEXS, Universitat Pompeu Fabra, Barcelona, Spain
| | - Andre M M Sousa
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, USA
| | - Nenad Sestan
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA.
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA.
- Yale Genome Editing Center, Yale School of Medicine, New Haven, CT, USA.
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA.
- Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA.
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT, USA.
- Kavli Institute for Neuroscience, Yale University, New Haven, CT, USA.
| |
Collapse
|
17
|
Moreau MX, Saillour Y, Cwetsch AW, Pierani A, Causeret F. Single-cell transcriptomics of the early developing mouse cerebral cortex disentangle the spatial and temporal components of neuronal fate acquisition. Development 2021; 148:269283. [PMID: 34170322 DOI: 10.1242/dev.197962] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 06/21/2021] [Indexed: 01/01/2023]
Abstract
In the developing cerebral cortex, how progenitors that seemingly display limited diversity end up producing a vast array of neurons remains a puzzling question. The prevailing model suggests that temporal maturation of progenitors is a key driver in the diversification of the neuronal output. However, temporal constraints are unlikely to account for all diversity, especially in the ventral and lateral pallium where neuronal types significantly differ from their dorsal neocortical counterparts born at the same time. In this study, we implemented single-cell RNAseq to sample the diversity of progenitors and neurons along the dorso-ventral axis of the early developing pallium. We first identified neuronal types, mapped them on the tissue and determined their origin through genetic tracing. We characterised progenitor diversity and disentangled the gene modules underlying temporal versus spatial regulations of neuronal specification. Finally, we reconstructed the developmental trajectories followed by ventral and dorsal pallial neurons to identify lineage-specific gene waves. Our data suggest a model by which discrete neuronal fate acquisition from a continuous gradient of progenitors results from the superimposition of spatial information and temporal maturation.
Collapse
Affiliation(s)
- Matthieu X Moreau
- Université de Paris, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, F-75015, Paris, France.,Université de Paris, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, F-75014, Paris, France
| | - Yoann Saillour
- Université de Paris, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, F-75015, Paris, France.,Université de Paris, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, F-75014, Paris, France
| | - Andrzej W Cwetsch
- Université de Paris, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, F-75015, Paris, France.,Université de Paris, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, F-75014, Paris, France
| | - Alessandra Pierani
- Université de Paris, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, F-75015, Paris, France.,Université de Paris, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, F-75014, Paris, France
| | - Frédéric Causeret
- Université de Paris, Imagine Institute, Team Genetics and Development of the Cerebral Cortex, F-75015, Paris, France.,Université de Paris, Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, F-75014, Paris, France
| |
Collapse
|
18
|
Qin J, Wang M, Zhao T, Xiao X, Li X, Yang J, Yi L, Goffinet AM, Qu Y, Zhou L. Early Forebrain Neurons and Scaffold Fibers in Human Embryos. Cereb Cortex 2021; 30:913-928. [PMID: 31298263 DOI: 10.1093/cercor/bhz136] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/21/2019] [Accepted: 05/31/2019] [Indexed: 12/24/2022] Open
Abstract
Neural progenitor proliferation, neuronal migration, areal organization, and pioneer axon wiring are critical events during early forebrain development, yet remain incompletely understood, especially in human. Here, we studied forebrain development in human embryos aged 5 to 8 postconceptional weeks (WPC5-8), stages that correspond to the neuroepithelium/early marginal zone (WPC5), telencephalic preplate (WPC6 & 7), and incipient cortical plate (WPC8). We show that early telencephalic neurons are formed at the neuroepithelial stage; the most precocious ones originate from local telencephalic neuroepithelium and possibly from the olfactory placode. At the preplate stage, forebrain organization is quite similar in human and mouse in terms of areal organization and of differentiation of Cajal-Retzius cells, pioneer neurons, and axons. Like in mice, axons from pioneer neurons in prethalamus, ventral telencephalon, and cortical preplate cross the diencephalon-telencephalon junction and the pallial-subpallial boundary, forming scaffolds that could guide thalamic and cortical axons at later stages. In accord with this model, at the early cortical plate stage, corticofugal axons run in ventral telencephalon in close contact with scaffold neurons, which express CELSR3 and FZD3, two molecules that regulates formation of similar scaffolds in mice.
Collapse
Affiliation(s)
- Jingwen Qin
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Jinan University Guangzhou, P R China
| | - Meizhi Wang
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Jinan University Guangzhou, P R China
| | - Tianyun Zhao
- Department of Anesthesiology, Guangzhou Women and Children's Medical Center Guangzhou Medical University Guangzhou, P R China
| | - Xue Xiao
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Jinan University Guangzhou, P R China
| | - Xuejun Li
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Jinan University Guangzhou, P R China
| | - Jieping Yang
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center Guangzhou Medical University Guangzhou, P R China
| | - Lisha Yi
- Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center Guangzhou Medical University Guangzhou, P R China
| | - Andre M Goffinet
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Jinan University Guangzhou, P R China
| | - Yibo Qu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Jinan University Guangzhou, P R China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory Guangzhou, P R China
| | - Libing Zhou
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory Jinan University Guangzhou, P R China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory Guangzhou, P R China.,Key Laboratory of Neuroscience, School of Basic Medical Sciences; Institute of Neuroscience, The Second Affiliated Hospital Guangzhou Medical University Guangzhou, P R China
| |
Collapse
|
19
|
Vasistha NA, Khodosevich K. The impact of (ab)normal maternal environment on cortical development. Prog Neurobiol 2021; 202:102054. [PMID: 33905709 DOI: 10.1016/j.pneurobio.2021.102054] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/01/2021] [Accepted: 04/20/2021] [Indexed: 12/24/2022]
Abstract
The cortex in the mammalian brain is the most complex brain region that integrates sensory information and coordinates motor and cognitive processes. To perform such functions, the cortex contains multiple subtypes of neurons that are generated during embryogenesis. Newly born neurons migrate to their proper location in the cortex, grow axons and dendrites, and form neuronal circuits. These developmental processes in the fetal brain are regulated to a large extent by a great variety of factors derived from the mother - starting from simple nutrients as building blocks and ending with hormones. Thus, when the normal maternal environment is disturbed due to maternal infection, stress, malnutrition, or toxic substances, it might have a profound impact on cortical development and the offspring can develop a variety of neurodevelopmental disorders. Here we first describe the major developmental processes which generate neuronal diversity in the cortex. We then review our knowledge of how most common maternal insults affect cortical development, perturb neuronal circuits, and lead to neurodevelopmental disorders. We further present a concept of selective vulnerability of cortical neuronal subtypes to maternal-derived insults, where the vulnerability of cortical neurons and their progenitors to an insult depends on the time (developmental period), place (location in the developing brain), and type (unique features of a cell type and an insult). Finally, we provide evidence for the existence of selective vulnerability during cortical development and identify the most vulnerable neuronal types, stages of differentiation, and developmental time for major maternal-derived insults.
Collapse
Affiliation(s)
- Navneet A Vasistha
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.
| | - Konstantin Khodosevich
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.
| |
Collapse
|
20
|
Changeux JP, Goulas A, Hilgetag CC. A Connectomic Hypothesis for the Hominization of the Brain. Cereb Cortex 2021; 31:2425-2449. [PMID: 33367521 PMCID: PMC8023825 DOI: 10.1093/cercor/bhaa365] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/30/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023] Open
Abstract
Cognitive abilities of the human brain, including language, have expanded dramatically in the course of our recent evolution from nonhuman primates, despite only minor apparent changes at the gene level. The hypothesis we propose for this paradox relies upon fundamental features of human brain connectivity, which contribute to a characteristic anatomical, functional, and computational neural phenotype, offering a parsimonious framework for connectomic changes taking place upon the human-specific evolution of the genome. Many human connectomic features might be accounted for by substantially increased brain size within the global neural architecture of the primate brain, resulting in a larger number of neurons and areas and the sparsification, increased modularity, and laminar differentiation of cortical connections. The combination of these features with the developmental expansion of upper cortical layers, prolonged postnatal brain development, and multiplied nongenetic interactions with the physical, social, and cultural environment gives rise to categorically human-specific cognitive abilities including the recursivity of language. Thus, a small set of genetic regulatory events affecting quantitative gene expression may plausibly account for the origins of human brain connectivity and cognition.
Collapse
Affiliation(s)
- Jean-Pierre Changeux
- CNRS UMR 3571, Institut Pasteur, 75724 Paris, France
- Communications Cellulaires, Collège de France, 75005 Paris, France
| | - Alexandros Goulas
- Institute of Computational Neuroscience, University Medical Center Eppendorf, Hamburg University, 20246 Hamburg, Germany
| | - Claus C Hilgetag
- Institute of Computational Neuroscience, University Medical Center Eppendorf, Hamburg University, 20246 Hamburg, Germany
- Department of Health Sciences, Boston University, Boston, MA 02115, USA
| |
Collapse
|
21
|
Homman-Ludiye J, Bourne JA. The Marmoset: The Next Frontier in Understanding the Development of the Human Brain. ILAR J 2021; 61:248-259. [PMID: 33620074 DOI: 10.1093/ilar/ilaa028] [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] [Received: 02/27/2020] [Revised: 11/13/2020] [Accepted: 11/20/2020] [Indexed: 12/22/2022] Open
Abstract
Rodent models, particularly mice, have dominated the field of developmental neuroscience for decades, like they have in most fields of biomedicine research. However, with 80 million years since rodents and primates last shared a common ancestor, the use of mice to model the development of the human brain is not without many shortcomings. The human brain diverges from the mouse brain in many aspects and is comprised of novel structures as well as diversified cellular subtypes. While these newly evolved features have no equivalent in rodents, they are observed in nonhuman primates. Therefore, elucidating the cellular mechanisms underlying the development and maturation of the healthy and diseased human brain can be achieved using less complex nonhuman primates. Historically, macaques were the preferred nonhuman primate model. However, over the past decade, the New World marmoset monkey (Callithrix jacchus) has gained more importance, particularly in the field of neurodevelopment. With its small size, twin or triplet birth, and prosocial behavior, the marmoset is an ideal model to study normal brain development as well as neurodevelopmental disorders, which are often associated with abnormal social behaviors. The growing interest in the marmoset has prompted many comparative studies, all demonstrating that the marmoset brain closely resembles that of the human and is perfectly suited to model human brain development. The marmoset is thus poised to extend its influence in the field of neurodevelopment and will hopefully fill the gaps that the mouse has left in our understanding of how our brain forms and how neurodevelopmental disorders originate.
Collapse
Affiliation(s)
- Jihane Homman-Ludiye
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - James A Bourne
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
22
|
Xu L, Zheng Y, Li X, Wang A, Huo D, Li Q, Wang S, Luo Z, Liu Y, Xu F, Wu X, Wu M, Zhou Y. Abnormal neocortex arealization and Sotos-like syndrome-associated behavior in Setd2 mutant mice. SCIENCE ADVANCES 2021; 7:7/1/eaba1180. [PMID: 33523829 PMCID: PMC7775761 DOI: 10.1126/sciadv.aba1180] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 11/11/2020] [Indexed: 06/12/2023]
Abstract
Proper formation of area identities of the cerebral cortex is crucial for cognitive functions and social behaviors of the brain. It remains largely unknown whether epigenetic mechanisms, including histone methylation, regulate cortical arealization. Here, we removed SETD2, the methyltransferase for histone 3 lysine-36 trimethylation (H3K36me3), in the developing dorsal forebrain in mice and showed that Setd2 is required for proper cortical arealization and the formation of cortico-thalamo-cortical circuits. Moreover, Setd2 conditional knockout mice exhibit defects in social interaction, motor learning, and spatial memory, reminiscent of patients with the Sotos-like syndrome bearing SETD2 mutations. SETD2 maintains the expression of clustered protocadherin (cPcdh) genes in an H3K36me3 methyltransferase-dependent manner. Aberrant cortical arealization was recapitulated in cPcdh heterozygous mice. Together, our study emphasizes epigenetic mechanisms underlying cortical arealization and pathogenesis of the Sotos-like syndrome.
Collapse
Affiliation(s)
- Lichao Xu
- College of Life Sciences, Department of Neurosurgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, and Medical Research Institute at School of Medicine, Wuhan University, Wuhan 430071, China
| | - Yue Zheng
- College of Life Sciences, Department of Neurosurgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, and Medical Research Institute at School of Medicine, Wuhan University, Wuhan 430071, China
| | - Xuejing Li
- College of Life Sciences, Department of Neurosurgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, and Medical Research Institute at School of Medicine, Wuhan University, Wuhan 430071, China
| | - Andi Wang
- College of Life Sciences, Department of Neurosurgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, and Medical Research Institute at School of Medicine, Wuhan University, Wuhan 430071, China
| | - Dawei Huo
- Department of Cell Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Qixiangtai Road 22, Tianjin 300070, China
- Department of Neurosurgery, Tianjin Medical University General Hospital and Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin 300052, China
| | - Qinglan Li
- College of Life Sciences, Department of Neurosurgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, and Medical Research Institute at School of Medicine, Wuhan University, Wuhan 430071, China
| | - Shikang Wang
- College of Life Sciences, Department of Neurosurgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, and Medical Research Institute at School of Medicine, Wuhan University, Wuhan 430071, China
| | - Zhiyuan Luo
- College of Life Sciences, Department of Neurosurgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, and Medical Research Institute at School of Medicine, Wuhan University, Wuhan 430071, China
| | - Ying Liu
- College of Life Sciences, Department of Neurosurgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, and Medical Research Institute at School of Medicine, Wuhan University, Wuhan 430071, China
| | - Fuqiang Xu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China
| | - Xudong Wu
- Department of Cell Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Qixiangtai Road 22, Tianjin 300070, China
- Department of Neurosurgery, Tianjin Medical University General Hospital and Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin 300052, China
| | - Min Wu
- College of Life Sciences, Department of Neurosurgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, and Medical Research Institute at School of Medicine, Wuhan University, Wuhan 430071, China.
| | - Yan Zhou
- College of Life Sciences, Department of Neurosurgery, Zhongnan Hospital of Wuhan University; Frontier Science Center for Immunology and Metabolism, and Medical Research Institute at School of Medicine, Wuhan University, Wuhan 430071, China.
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
| |
Collapse
|
23
|
Ratzan EM, Moon AM, Deans MR. Fgf8 genetic labeling reveals the early specification of vestibular hair cell type in mouse utricle. Development 2020; 147:dev.192849. [PMID: 33046506 DOI: 10.1242/dev.192849] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 10/07/2020] [Indexed: 01/16/2023]
Abstract
FGF8 signaling plays diverse roles in inner ear development, acting at multiple stages from otic placode induction to cellular differentiation in the organ of Corti. As a secreted morphogen with diverse functions, Fgf8 expression is likely to be spatially restricted and temporally dynamic throughout inner ear development. We evaluated these characteristics using genetic labeling mediated by Fgf8 mcm gene-targeted mice and determined that Fgf8 expression is a specific and early marker of Type-I vestibular hair cell identity. Fgf8 mcm expression initiates at E11.5 in the future striolar region of the utricle, labeling hair cells following EdU birthdating, and demonstrates that sub-type identity is determined shortly after terminal mitosis. This early fate specification is not apparent using markers or morphological criteria that are not present before birth in the mouse. Although analyses of Fgf8 conditional knockout mice did not reveal developmental phenotypes, the restricted pattern of Fgf8 expression suggests that functionally redundant FGF ligands may contribute to vestibular hair cell differentiation and supports a developmental model in which Type-I and Type-II hair cells develop in parallel rather than from an intermediate precursor.
Collapse
Affiliation(s)
- Evan M Ratzan
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA.,Interdepartmental Program in Neuroscience, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Anne M Moon
- Departments of Molecular and Functional Genomics and Pediatrics, Weis Center for Research, Geisinger Clinic and Geisinger Commonwealth School of Medicine, Danville, PA 17822, USA.,Departments of Pediatrics and Human Genetics, University of Utah, Salt Lake City, UT 84112 USA
| | - Michael R Deans
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA .,Department of Surgery, Division of Otolaryngology, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| |
Collapse
|
24
|
Self-organization of cortical areas in the development and evolution of neocortex. Proc Natl Acad Sci U S A 2020; 117:29212-29220. [PMID: 33139564 DOI: 10.1073/pnas.2011724117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
While the mechanisms generating the topographic organization of primary sensory areas in the neocortex are well studied, what generates secondary cortical areas is virtually unknown. Using physical parameters representing primary and secondary visual areas as they vary from monkey to mouse, we derived a network growth model to explore if characteristic features of secondary areas could be produced from correlated activity patterns arising from V1 alone. We found that V1 seeded variable numbers of secondary areas based on activity-driven wiring and wiring-density limits within the cortical surface. These secondary areas exhibited the typical mirror-reversal of map topography on cortical area boundaries and progressive reduction of the area and spatial resolution of each new map on the caudorostral axis. Activity-based map formation may be the basic mechanism that establishes the matrix of topographically organized cortical areas available for later computational specialization.
Collapse
|
25
|
García-Moreno F, Molnár Z. Variations of telencephalic development that paved the way for neocortical evolution. Prog Neurobiol 2020; 194:101865. [PMID: 32526253 PMCID: PMC7656292 DOI: 10.1016/j.pneurobio.2020.101865] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/29/2020] [Accepted: 06/05/2020] [Indexed: 12/13/2022]
Abstract
Charles Darwin stated, "community in embryonic structure reveals community of descent". Thus, to understand how the neocortex emerged during mammalian evolution we need to understand the evolution of the development of the pallium, the source of the neocortex. In this article, we review the variations in the development of the pallium that enabled the production of the six-layered neocortex. We propose that an accumulation of subtle modifications from very early brain development accounted for the diversification of vertebrate pallia and the origin of the neocortex. Initially, faint differences of expression of secretable morphogens promote a wide variety in the proportions and organization of sectors of the early pallium in different vertebrates. It prompted different sectors to host varied progenitors and distinct germinative zones. These cells and germinative compartments generate diverse neuronal populations that migrate and mix with each other through radial and tangential migrations in a taxon-specific fashion. Together, these early variations had a profound influence on neurogenetic gradients, lamination, positioning, and connectivity. Gene expression, hodology, and physiological properties of pallial neurons are important features to suggest homologies, but the origin of cells and their developmental trajectory are fundamental to understand evolutionary changes. Our review compares the development of the homologous pallial sectors in sauropsids and mammals, with a particular focus on cell lineage, in search of the key changes that led to the appearance of the mammalian neocortex.
Collapse
Affiliation(s)
- Fernando García-Moreno
- Achucarro Basque Center for Neuroscience, Scientific Park of the University of the Basque Country (UPV/EHU), 48940, Leioa, Spain; IKERBASQUE Foundation, María Díaz de Haro 3, 6th Floor, 48013, Bilbao, Spain; Department of Neuroscience, Faculty of Medicine and Odontology, UPV/EHU, Barrio Sarriena s/n, 48940, Leioa, Bizkaia, Spain.
| | - Zoltán Molnár
- Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Oxford, OX1 3QX, UK.
| |
Collapse
|
26
|
Di Nardo AA, Joliot A, Prochiantz A. Homeoprotein transduction in neurodevelopment and physiopathology. SCIENCE ADVANCES 2020; 6:6/44/eabc6374. [PMID: 33115744 PMCID: PMC7608782 DOI: 10.1126/sciadv.abc6374] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 09/11/2020] [Indexed: 05/28/2023]
Abstract
Homeoproteins were originally identified for embryonic cell-autonomous transcription activity, but they also have non-cell-autonomous activity owing to transfer between cells. This Review discusses transfer mechanisms and focuses on some established functions, such as neurodevelopmental regulation of axon guidance, and postnatal critical periods of brain plasticity that affect sensory processing and cognition. Homeoproteins are present across all eukaryotes, and intercellular transfer occurs in plants and animals. Proposed functions have evolutionary relevance, such as morphogenetic activity and sexual exchange during the mating of unicellular eukaryotes, while others have physiopathological relevance, such as regulation of mood and cognition by influencing brain compartmentalization, connectivity, and plasticity. There are more than 250 known homeoproteins with conserved transfer domains, suggesting that this is a common mode of signal transduction but with many undiscovered functions.
Collapse
Affiliation(s)
- Ariel A Di Nardo
- Centre for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR 7241, INSERM U1050, PSL University, Labex MemoLife, 75005 Paris, France.
| | - Alain Joliot
- Centre for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR 7241, INSERM U1050, PSL University, Labex MemoLife, 75005 Paris, France
| | - Alain Prochiantz
- Centre for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR 7241, INSERM U1050, PSL University, Labex MemoLife, 75005 Paris, France.
| |
Collapse
|
27
|
Kostović I. The enigmatic fetal subplate compartment forms an early tangential cortical nexus and provides the framework for construction of cortical connectivity. Prog Neurobiol 2020; 194:101883. [PMID: 32659318 DOI: 10.1016/j.pneurobio.2020.101883] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 06/05/2020] [Accepted: 07/06/2020] [Indexed: 12/19/2022]
Abstract
The most prominent transient compartment of the primate fetal cortex is the deep, cell-sparse, synapse-containing subplate compartment (SPC). The developmental role of the SPC and its extraordinary size in humans remain enigmatic. This paper evaluates evidence on the development and connectivity of the SPC and discusses its role in the pathogenesis of neurodevelopmental disorders. A synthesis of data shows that the subplate becomes a prominent compartment by its expansion from the deep cortical plate (CP), appearing well-delineated on MR scans and forming a tangential nexus across the hemisphere, consisting of an extracellular matrix, randomly distributed postmigratory neurons, multiple branches of thalamic and long corticocortical axons. The SPC generates early spontaneous non-synaptic and synaptic activity and mediates cortical response upon thalamic stimulation. The subplate nexus provides large-scale interareal connectivity possibly underlying fMR resting-state activity, before corticocortical pathways are established. In late fetal phase, when synapses appear within the CP, transient the SPC coexists with permanent circuitry. The histogenetic role of the SPC is to provide interactive milieu and capacity for guidance, sorting, "waiting" and target selection of thalamocortical and corticocortical pathways. The new evolutionary role of the SPC and its remnant white matter neurons is linked to the increasing number of associative pathways in the human neocortex. These roles attributed to the SPC are regulated using a spatiotemporal gene expression during critical periods, when pathogenic factors may disturb vulnerable circuitry of the SPC, causing neurodevelopmental cognitive circuitry disorders.
Collapse
Affiliation(s)
- Ivica Kostović
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, Salata 12, 10000 Zagreb, Croatia.
| |
Collapse
|
28
|
Pattabiraman K, Muchnik SK, Sestan N. The evolution of the human brain and disease susceptibility. Curr Opin Genet Dev 2020; 65:91-97. [PMID: 32629339 DOI: 10.1016/j.gde.2020.05.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/05/2020] [Accepted: 05/01/2020] [Indexed: 12/13/2022]
Abstract
Evolutionary perspective is critical for understanding human biology, human medicine, and the traits that make human beings unique. One of the crucial characteristics that sets humans apart from other extant species is our cognitive ability, which allows for complex processes including symbolic thought, theory of mind, and syntactical-grammatical language, and is thought to arise from the expansion and specialization of the human nervous system. It has been hypothesized that the same evolutionary changes that allowed us to develop these valuable skills made humans susceptible to neurodevelopmental and neurodegenerative disease. Unfortunately, our lack of access to our extinct ancestors makes this a difficult hypothesis to test, but recent collaborations between the fields of evolution, genetics, genomics, neuroscience, neurology and psychiatry have begun to provide some clues. Here, we will outline recent work in those fields that have utilized our growing knowledge of disease risk genes and loci, identified by wide-scale genetic studies, and nervous system development and function to draw conclusions about the impact of human-specific aspects of evolution. We will discuss studies that assess evolution at a variety of scales including at the levels of whole brain regions, cell types, synapses, metabolic processes, gene expression patterns, and gene regulation. At all of these levels, there is preliminary evidence that human-specific brain features are linked to neurodevelopmental and neurodegenerative disease risk.
Collapse
Affiliation(s)
- Kartik Pattabiraman
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Yale Child Study Center, New Haven, CT 06510, USA
| | - Sydney Keaton Muchnik
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Nenad Sestan
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Yale Child Study Center, New Haven, CT 06510, USA; Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA; Departments of Psychiatry and Comparative Medicine, Kavli Institute for Neuroscience, Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
29
|
Del Pino I, Tocco C, Magrinelli E, Marcantoni A, Ferraguto C, Tomagra G, Bertacchi M, Alfano C, Leinekugel X, Frick A, Studer M. COUP-TFI/Nr2f1 Orchestrates Intrinsic Neuronal Activity during Development of the Somatosensory Cortex. Cereb Cortex 2020; 30:5667-5685. [DOI: 10.1093/cercor/bhaa137] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/17/2020] [Accepted: 04/29/2020] [Indexed: 01/19/2023] Open
Abstract
Abstract
The formation of functional cortical maps in the cerebral cortex results from a timely regulated interaction between intrinsic genetic mechanisms and electrical activity. To understand how transcriptional regulation influences network activity and neuronal excitability within the neocortex, we used mice deficient for Nr2f1 (also known as COUP-TFI), a key determinant of primary somatosensory (S1) area specification during development. We found that the cortical loss of Nr2f1 impacts on spontaneous network activity and synchronization of S1 cortex at perinatal stages. In addition, we observed alterations in the intrinsic excitability and morphological features of layer V pyramidal neurons. Accordingly, we identified distinct voltage-gated ion channels regulated by Nr2f1 that might directly influence intrinsic bioelectrical properties during critical time windows of S1 cortex specification. Altogether, our data suggest a tight link between Nr2f1 and neuronal excitability in the developmental sequence that ultimately sculpts the emergence of cortical network activity within the immature neocortex.
Collapse
Affiliation(s)
- Isabel Del Pino
- Université de Bordeaux, Inserm U1215, Neurocentre Magendie, 33077 Bordeaux, France
- Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain
| | - Chiara Tocco
- Université Côte d’Azur, CNRS, Inserm, iBV, 06108 Nice, France
| | - Elia Magrinelli
- Université Côte d’Azur, CNRS, Inserm, iBV, 06108 Nice, France
- Département des Neurosciences Fondamentales, Université de Lausanne, CH-1005 Lausanne, Switzerland
| | - Andrea Marcantoni
- Dipartimento di Scienza e Tecnologia del Farmaco, Università di Torino, 10125 Torino, Italy
| | | | - Giulia Tomagra
- Dipartimento di Scienza e Tecnologia del Farmaco, Università di Torino, 10125 Torino, Italy
| | | | | | - Xavier Leinekugel
- Université de Bordeaux, Inserm U1215, Neurocentre Magendie, 33077 Bordeaux, France
| | - Andreas Frick
- Université de Bordeaux, Inserm U1215, Neurocentre Magendie, 33077 Bordeaux, France
| | - Michèle Studer
- Université Côte d’Azur, CNRS, Inserm, iBV, 06108 Nice, France
| |
Collapse
|
30
|
Khozhai LI, Otellin VA. Long-Term Effects of Perinatal Hypoxia on the Distribution of GABAergic Neurons in the Rat Neocortex. J EVOL BIOCHEM PHYS+ 2019. [DOI: 10.1134/s0022093019040100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
31
|
Klatt A, Salzmann E, Schneider LJ, Reifschneider A, Korneck M, Hermle P, Bürkle A, Stoll D, Kadereit S. Toxicity of ionizing radiation (IR) in a human induced pluripotent stem cell (hiPSC)-derived 3D early neurodevelopmental model. Arch Toxicol 2019; 93:2879-2893. [DOI: 10.1007/s00204-019-02553-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 08/22/2019] [Indexed: 01/04/2023]
|
32
|
Jones WD, Guadiana SM, Grove EA. A model of neocortical area patterning in the lissencephalic mouse may hold for larger gyrencephalic brains. J Comp Neurol 2019; 527:1461-1477. [PMID: 30689213 DOI: 10.1002/cne.24643] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/21/2018] [Accepted: 01/02/2019] [Indexed: 12/14/2022]
Abstract
In the mouse, two telencephalic signaling centers orchestrate embryonic patterning of the cerebral cortex. From the rostral patterning center in the telencephalon, the Fibroblast Growth Factor, FGF8, disperses as a morphogen to establish the rostral to caudal axis of the neocortical area map. FGF8 coordinates with Wnt3a from the cortical hem to regulate graded expression of transcription factors that position neocortical areas, and control hippocampal development. Whether similar signaling centers pattern the much larger cortices of carnivore and primate species, however, is unclear. The limited dispersion range of FGF8 and Wnt3a is inconsistent with patterning larger cortical primordia. Yet the implication that different mechanisms organize cortex in different mammals flies in the face of the tenet that developmental patterning mechanisms are conserved across vertebrate species. In the present study, both signaling centers were identified in the ferret telencephalon, as were expression gradients of the patterning transcription factor genes regulated by FGF8 and Wnt3a. Notably, at the stage corresponding to the peak period of FGF8 signaling in the mouse neocortical primordium (NP), the NP was the same size in ferret and mouse, which would allow morphogen patterning of the ferret NP. Subsequently, the size of ferret neocortex shot past that of the mouse. Images from online databases further suggest that NP growth in humans, too, is slowed in early cortical development. We propose that if early growth in larger brains is held back, mechanisms that pattern the neocortical area map in the mouse could be conserved across mammalian species.
Collapse
Affiliation(s)
- William D Jones
- Department of Neurobiology, University of Chicago, Chicago, Illinois
| | - Sarah M Guadiana
- Department of Neurobiology, University of Chicago, Chicago, Illinois
| | - Elizabeth A Grove
- Department of Neurobiology, University of Chicago, Chicago, Illinois.,Committee on Development, Regeneration and Stem Cell Biology, University of Chicago, Chicago, Illinois.,Committee on Neurobiology, University of Chicago, Chicago, Illinois
| |
Collapse
|
33
|
Martínez-Martínez MÁ, Ciceri G, Espinós A, Fernández V, Marín O, Borrell V. Extensive branching of radially-migrating neurons in the mammalian cerebral cortex. J Comp Neurol 2019; 527:1558-1576. [PMID: 30520050 DOI: 10.1002/cne.24597] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 11/22/2018] [Accepted: 11/28/2018] [Indexed: 11/06/2022]
Abstract
Excitatory neurons of the cerebral cortex migrate radially from their place of birth to their final position in the cortical plate during development. Radially-migrating neurons display a single leading process that establishes the direction of movement. This leading process has been described as being unbranched, and the occurrence of branches proposed to impair radial migration. Here we have analyzed the detailed morphology of leading process in radially-migrating pyramidal neurons and its impact on radial migration. We have compared ferret and mouse to identify differences between cortices that undergo folding or not. In mouse, we find that half of radially-migrating neurons exhibit a branched leading process, this being even more frequent in ferret. Branched leading processes are less parallel to radial glia fibers than those unbranched, suggesting some independence from radial glia fibers. Two-photon videomicroscopy revealed that a vast majority of neurons branch their leading process at some point during radial migration, but this does not reduce their migration speed. We have tested the functional impact of exuberant leading process branching by expressing a dominant negative Cdk5. We confirm that loss of Cdk5 function significantly impairs radial migration, but this is independent from increased branching of the leading process. We propose that excitatory neurons may branch their leading process as an evolutionary mechanism to allow cells changing their trajectory of migration to disperse laterally, such that increased branching in gyrencephalic species favors the tangential dispersion of radially-migrating neurons, and cortical folding.
Collapse
Affiliation(s)
- Maria Á Martínez-Martínez
- Instituto de Neurociencias, Agencia Estatal Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, San Juan de Alicante, Spain
| | - Gabriele Ciceri
- Instituto de Neurociencias, Agencia Estatal Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, San Juan de Alicante, Spain
| | - Alexandre Espinós
- Instituto de Neurociencias, Agencia Estatal Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, San Juan de Alicante, Spain
| | - Virginia Fernández
- Instituto de Neurociencias, Agencia Estatal Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, San Juan de Alicante, Spain
| | - Oscar Marín
- Instituto de Neurociencias, Agencia Estatal Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, San Juan de Alicante, Spain.,Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| | - Víctor Borrell
- Instituto de Neurociencias, Agencia Estatal Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, San Juan de Alicante, Spain
| |
Collapse
|
34
|
Liester MB, Sullivan EE. A review of epigenetics in human consciousness. COGENT PSYCHOLOGY 2019. [DOI: 10.1080/23311908.2019.1668222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Affiliation(s)
- Mitchell B. Liester
- Department of Psychiatry, University of Colorado School of Medicine, P.O. Box 302 153 N. Washington Street, Suite 103, Monument, CO 80132, USA
| | - Erin E. Sullivan
- Computer Science, University of Oklahoma, P.O. Box 302, Monument, CO 80132, USA
| |
Collapse
|
35
|
Holley ZL, Bland KM, Casey ZO, Handwerk CJ, Vidal GS. Cross-Regional Gradient of Dendritic Morphology in Isochronically-Sourced Mouse Supragranular Pyramidal Neurons. Front Neuroanat 2018; 12:103. [PMID: 30564104 PMCID: PMC6288488 DOI: 10.3389/fnana.2018.00103] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 11/15/2018] [Indexed: 11/13/2022] Open
Abstract
Architectonic heterogeneity in neurons is thought to be important for equipping the mammalian cerebral cortex with an adaptable network that can organize the manifold totality of information it receives. To this end, the dendritic arbors of supragranular pyramidal neurons, even those of the same class, are known to vary substantially. This diversity of dendritic morphology appears to have a rostrocaudal configuration in some brain regions of various species. For example, in humans and non-human primates, neurons in more rostral visual association areas (e.g., V4) tend to have more complex dendritic arbors than those in the caudal primary visual cortex. A rostrocaudal configuration is not so clear in any region of the mouse, which is increasingly being used as a model for neurodevelopmental disorders that arise from dysfunctional cerebral cortical circuits. Therefore, in this study we investigated the complexity of dendritic arbors of neurons distributed throughout a broad area of the mouse cerebral cortex. We reduced selection bias by labeling neurons restricted to become supragranular pyramidal neurons using in utero electroporation. While we observed that the simple rostrocaudal position, cortical depth, or even functional region of a neuron was not directly related to its dendritic morphology, a model that instead included a caudomedial-to-rostrolateral gradient accounted for a significant amount of the observed dendritic morphological variance. In other words, rostrolateral neurons from our data set were generally more complex when compared to caudomedial neurons. Furthermore, dividing the cortex into a visual area and a non-visual area maintained the power of the relationship between caudomedial-to-rostrolateral position and dendritic complexity. Our observations therefore support the idea that dendritic morphology of mouse supragranular excitatory pyramidal neurons across much of the tangential plane of the cerebral cortex is partly shaped by a developmental gradient spanning several functional regions.
Collapse
Affiliation(s)
- Zachary Logan Holley
- Department of Biology, James Madison University, Harrisonburg, VA, United States
| | - Katherine M Bland
- Department of Biology, James Madison University, Harrisonburg, VA, United States
| | - Zachary O Casey
- Department of Biology, James Madison University, Harrisonburg, VA, United States
| | | | - George S Vidal
- Department of Biology, James Madison University, Harrisonburg, VA, United States
| |
Collapse
|
36
|
Krubitzer LA, Prescott TJ. The Combinatorial Creature: Cortical Phenotypes within and across Lifetimes. Trends Neurosci 2018; 41:744-762. [PMID: 30274608 DOI: 10.1016/j.tins.2018.08.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 07/31/2018] [Accepted: 08/02/2018] [Indexed: 12/15/2022]
Abstract
The neocortex is one of the most distinctive structures of the mammalian brain, yet also one of the most varied in terms of both size and organization. Multiple processes have contributed to this variability, including evolutionary mechanisms (i.e., alterations in gene sequence) that alter the size, organization, and connections of neocortex, and activity dependent mechanisms that can also modify these same features. Thus, changes to the neocortex can occur over different time-scales, including within a single generation. This combination of genetic and activity dependent mechanisms that create a given cortical phenotype allows the mammalian neocortex to rapidly and flexibly adjust to different body and environmental contexts, and in humans permits culture to impact brain construction.
Collapse
Affiliation(s)
- Leah A Krubitzer
- Center for Neuroscience and Department of Psychology, University of California, Davis, Davis, CA 95616, USA.
| | - Tony J Prescott
- Sheffield Robotics and Department of Computer Science, University of Sheffield, Sheffield, UK
| |
Collapse
|
37
|
Vieira MS, Santos AK, Vasconcellos R, Goulart VAM, Parreira RC, Kihara AH, Ulrich H, Resende RR. Neural stem cell differentiation into mature neurons: Mechanisms of regulation and biotechnological applications. Biotechnol Adv 2018; 36:1946-1970. [PMID: 30077716 DOI: 10.1016/j.biotechadv.2018.08.002] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 07/31/2018] [Accepted: 08/01/2018] [Indexed: 02/07/2023]
Abstract
The abilities of stem cells to self-renew and form different mature cells expand the possibilities of applications in cell-based therapies such as tissue recomposition in regenerative medicine, drug screening, and treatment of neurodegenerative diseases. In addition to stem cells found in the embryo, various adult organs and tissues have niches of stem cells in an undifferentiated state. In the central nervous system of adult mammals, neurogenesis occurs in two regions: the subventricular zone and the dentate gyrus in the hippocampus. The generation of the different neural lines originates in adult neural stem cells that can self-renew or differentiate into astrocytes, oligodendrocytes, or neurons in response to specific stimuli. The regulation of the fate of neural stem cells is a finely controlled process relying on a complex regulatory network that extends from the epigenetic to the translational level and involves extracellular matrix components. Thus, a better understanding of the mechanisms underlying how the process of neurogenesis is induced, regulated, and maintained will provide elues for development of novel for strategies for neurodegenerative therapies. In this review, we focus on describing the mechanisms underlying the regulation of the neuronal differentiation process by transcription factors, microRNAs, and extracellular matrix components.
Collapse
Affiliation(s)
- Mariana S Vieira
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nanocell, Divinopólis, MG, Brazil
| | - Anderson K Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rebecca Vasconcellos
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nanocell, Divinopólis, MG, Brazil
| | - Vânia A M Goulart
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ricardo C Parreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nanocell, Divinopólis, MG, Brazil
| | - Alexandre H Kihara
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, SP, Brazil.
| | - Rodrigo R Resende
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nanocell, Divinopólis, MG, Brazil.
| |
Collapse
|
38
|
Bertacchi M, Parisot J, Studer M. The pleiotropic transcriptional regulator COUP-TFI plays multiple roles in neural development and disease. Brain Res 2018; 1705:75-94. [PMID: 29709504 DOI: 10.1016/j.brainres.2018.04.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 04/19/2018] [Accepted: 04/20/2018] [Indexed: 12/23/2022]
Abstract
Transcription factors are expressed in a dynamic fashion both in time and space during brain development, and exert their roles by activating a cascade of multiple target genes. This implies that understanding the precise function of a transcription factor becomes a challenging task. In this review, we will focus on COUP-TFI (or NR2F1), a nuclear receptor belonging to the superfamily of the steroid/thyroid hormone receptors, and considered to be one of the major transcriptional regulators orchestrating cortical arealization, cell-type specification and maturation. Recent data have unraveled the multi-faceted functions of COUP-TFI in the development of several mouse brain structures, including the neocortex, hippocampus and ganglionic eminences. Despite NR2F1 mutations and deletions in humans have been linked to a complex neurodevelopmental disease mainly associated to optic atrophy and intellectual disability, its role during the formation of the retina and optic nerve remains unclear. In light of its major influence in cortical development, we predict that its haploinsufficiency might be the cause of other cognitive diseases, not identified so far. Mouse models offer a unique opportunity of dissecting COUP-TFI function in different regions during brain assembly; hence, the importance of comparing and discussing common points linking mouse models to human patients' symptoms.
Collapse
Affiliation(s)
- Michele Bertacchi
- Université Côte d'Azur, CNRS, Inserm, iBV - Institut de Biologie Valrose, 06108 Nice, France.
| | - Josephine Parisot
- Université Côte d'Azur, CNRS, Inserm, iBV - Institut de Biologie Valrose, 06108 Nice, France
| | - Michèle Studer
- Université Côte d'Azur, CNRS, Inserm, iBV - Institut de Biologie Valrose, 06108 Nice, France.
| |
Collapse
|
39
|
Hébert JM, Vijg J. Cell Replacement to Reverse Brain Aging: Challenges, Pitfalls, and Opportunities. Trends Neurosci 2018; 41:267-279. [PMID: 29548515 DOI: 10.1016/j.tins.2018.02.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 01/23/2018] [Accepted: 02/12/2018] [Indexed: 12/21/2022]
Abstract
Current antiaging strategies focusing on druggable targets have met with relatively limited success to date. Replacement of cells, tissues, and organs could provide an alternative means for targeting age-induced damage and potentially eliminating some of it. However, before this is a viable option, numerous challenges need to be addressed. Most notably, whether the brain, which defines our self-identity, is amenable to replacement therapies is unclear. Here, we consider whether progressive cell replacement is a potential approach to reverse brain aging without grossly altering function. We focus mainly on the neocortex, seat of our highest cognitive functions, because of abundant knowledge on neocortical development, plasticity, and how the neocortex can functionally incorporate new neurons. We outline the primary challenges for brain cell replacement, and key areas that require further investigation.
Collapse
Affiliation(s)
- Jean M Hébert
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Jan Vijg
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
40
|
Adnani L, Han S, Li S, Mattar P, Schuurmans C. Mechanisms of Cortical Differentiation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 336:223-320. [DOI: 10.1016/bs.ircmb.2017.07.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
41
|
de Juan Romero C, Borrell V. Genetic maps and patterns of cerebral cortex folding. Curr Opin Cell Biol 2017; 49:31-37. [PMID: 29227862 DOI: 10.1016/j.ceb.2017.11.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 11/17/2017] [Accepted: 11/26/2017] [Indexed: 01/24/2023]
Abstract
Folding of the cerebral cortex during brain development is a complex process that depends on the orchestrated action of a number of factors, including generation and proliferation of basal progenitor cells, and the radial migration of neurons. Patterns of primary cortical folding are stereotyped between individuals and across phylogeny, reflecting a strong genetic regulation of the underlying cellular mechanisms. Here we summarize recent findings on cellular and genetic mechanisms regulating this fascinating process that underlies expansion and functional complexification of the mammalian cerebral cortex.
Collapse
Affiliation(s)
- Camino de Juan Romero
- Instituto de Neurociencias, Agencia Estatal Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, 03550 Sant Joan d'Alacant, Spain
| | - Víctor Borrell
- Instituto de Neurociencias, Agencia Estatal Consejo Superior de Investigaciones Científicas & Universidad Miguel Hernández, 03550 Sant Joan d'Alacant, Spain.
| |
Collapse
|
42
|
Yuzwa SA, Borrett MJ, Innes BT, Voronova A, Ketela T, Kaplan DR, Bader GD, Miller FD. Developmental Emergence of Adult Neural Stem Cells as Revealed by Single-Cell Transcriptional Profiling. Cell Rep 2017; 21:3970-3986. [DOI: 10.1016/j.celrep.2017.12.017] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 10/30/2017] [Accepted: 12/01/2017] [Indexed: 02/06/2023] Open
|
43
|
MANF Is Essential for Neurite Extension and Neuronal Migration in the Developing Cortex. eNeuro 2017; 4:eN-NWR-0214-17. [PMID: 29082311 PMCID: PMC5655607 DOI: 10.1523/eneuro.0214-17.2017] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 08/22/2017] [Accepted: 09/05/2017] [Indexed: 12/15/2022] Open
Abstract
Mesencephalic astrocyte-derived neurotrophic factor (MANF) is an endoplasmic reticulum (ER) resident protein with neuroprotective effects. Previous studies have shown that MANF expression is altered in the developing rodent cortex in a spatiotemporal manner. However, the role of MANF in mammalian neurogenesis is not known. The aim of this study was to determine the role of MANF in neural stem cell (NSC) proliferation, differentiation, and cerebral cortex development. We found that MANF is highly expressed in neural lineage cells, including NSCs in the developing brain. We discovered that MANF-deficient NSCs in culture are viable and show no defect in proliferation. However, MANF-deficient cells have deficits in neurite extension upon neuronal differentiation. In vivo, MANF removal leads to slower neuronal migration and impaired neurite outgrowth. In vitro, mechanistic studies indicate that impaired neurite growth is preceded by reduced de novo protein synthesis and constitutively activated unfolded protein response (UPR) pathways. This study is the first to demonstrate that MANF is a novel and critical regulator of neurite growth and neuronal migration in mammalian cortical development.
Collapse
|
44
|
Jabaudon D. Fate and freedom in developing neocortical circuits. Nat Commun 2017; 8:16042. [PMID: 28671189 PMCID: PMC5500875 DOI: 10.1038/ncomms16042] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 05/23/2017] [Indexed: 12/22/2022] Open
Abstract
The activity of neuronal circuits of the neocortex underlies our ability to perceive the world and interact with our environment. During development, these circuits emerge from dynamic interactions between cell-intrinsic, genetically determined programs and input/activity-dependent signals, which together shape these circuits into adulthood. Building on a large body of experimental work, several recent technological developments now allow us to interrogate these nature–nurture interactions with single gene/single input/single-cell resolution. Focusing on excitatory glutamatergic neurons, this review discusses the genetic and input-dependent mechanisms controlling how individual cortical neurons differentiate into specialized cells to assemble into stereotypical local circuits within global, large-scale networks.
Proper functioning of the neocortex – the center of higher-order brain functions – depends on the correct assembly of neocortical neural circuits during development. Here the author discusses how cell-intrinsic developmental programs and activity-dependent signals together shape the formation of neocortical circuits.
Collapse
Affiliation(s)
- Denis Jabaudon
- Department of Basic Neurosciences, Geneva University, 1 rue Michel Servet, 1211 Geneva, Switzerland.,Clinic of Neurology, Geneva University Hospital, 1 rue Michel Servet, 1211 Geneva, Switzerland.,Geneva Neurocenter, Geneva University, 1 rue Michel Servet, 1211 Geneva, Switzerland
| |
Collapse
|
45
|
Luhmann HJ, Khazipov R. Neuronal activity patterns in the developing barrel cortex. Neuroscience 2017; 368:256-267. [PMID: 28528963 DOI: 10.1016/j.neuroscience.2017.05.025] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/12/2017] [Accepted: 05/12/2017] [Indexed: 11/26/2022]
Abstract
The developing barrel cortex reveals a rich repertoire of neuronal activity patterns, which have been also found in other sensory neocortical areas and in other species including the somatosensory cortex of preterm human infants. The earliest stage is characterized by asynchronous, sparse single-cell firing at low frequencies. During the second stage neurons show correlated firing, which is initially mediated by electrical synapses and subsequently transforms into network bursts depending on chemical synapses. Activity patterns during this second stage are synchronous plateau assemblies, delta waves, spindle bursts and early gamma oscillations (EGOs). In newborn rodents spindle bursts and EGOs occur spontaneously or can be elicited by sensory stimulation and synchronize the activity in a barrel-related columnar network with topographic organization at the day of birth. Interfering with this early activity causes a disturbance in the development of the cortical architecture, indicating that spindle bursts and EGOs influence the formation of cortical columns. Early neuronal activity also controls the rate of programed cell death in the developing barrel cortex, suggesting that spindle bursts and EGOs are physiological activity patterns particularly suited to suppress apoptosis. It remains to be studied in more detail how these different neocortical activity patterns control early developmental processes such as formation of synapses, microcircuits, topographic maps and large-scale networks.
Collapse
Affiliation(s)
- Heiko J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University Mainz, Duesbergweg 6, D-55128 Mainz, Germany.
| | - Rustem Khazipov
- INMED - INSERM, Aix-Marseille University, Marseille 13273, France; Laboratory of Neurobiology, Kazan Federal University, Kazan 420008, Russia
| |
Collapse
|
46
|
Perkeybile AM, Bales KL. Intergenerational transmission of sociality: the role of parents in shaping social behavior in monogamous and non-monogamous species. J Exp Biol 2017; 220:114-123. [PMID: 28057834 PMCID: PMC5278619 DOI: 10.1242/jeb.142182] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Social bonds are necessary for many mammals to survive and reproduce successfully. These bonds (i.e. pair-bonds, friendships, filial bonds) are characterized by different periods of development, longevity and strength. Socially monogamous species display certain behaviors not seen in many other mammals, such as adult pair-bonding and male parenting. In our studies of prairie voles (Microtus ochrogaster) and titi monkeys (Callicebus cupreus), we have examined the neurohormonal basis of these bonds. Here, we discuss the evidence from voles that aspects of adolescent and adult social behavior are shaped by early experience, including changes to sensory systems and connections, neuropeptide systems such as oxytocin and vasopressin, and alterations in stress responses. We will compare this with what is known about these processes during development and adulthood in other mammalian species, both monogamous and non-monogamous, and how our current knowledge in voles can be used to understand the development of and variation in social bonds. Humans are endlessly fascinated by the variety of social relationships and family types displayed by animal species, including our own. Social relationships can be characterized by directionality (either uni- or bi-directional), longevity, developmental epoch (infant, juvenile or adult) and strength. Research on the neurobiology of social bonds in animals has focused primarily on 'socially monogamous' species, because of their long-term, strong adult affiliative bonds. In this Review, we attempt to understand how the ability and propensity to form these bonds (or lack thereof), as well as the display of social behaviors more generally, are transmitted both genomically and non-genomically via variation in parenting in monogamous and non-monogamous species.
Collapse
Affiliation(s)
- Allison M Perkeybile
- Department of Psychology, University of California, Davis, Davis, CA 95616, USA
- The Kinsey Institute, Indiana University, Bloomington, IN 47405, USA
| | - Karen L Bales
- Department of Psychology, University of California, Davis, Davis, CA 95616, USA
| |
Collapse
|
47
|
The marmoset: An emerging model to unravel the evolution and development of the primate neocortex. Dev Neurobiol 2016; 77:263-272. [DOI: 10.1002/dneu.22425] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 07/25/2016] [Accepted: 07/29/2016] [Indexed: 12/13/2022]
|
48
|
Chen X, Petit EI, Dobrenis K, Sze JY. Spatiotemporal SERT expression in cortical map development. Neurochem Int 2016; 98:129-37. [PMID: 27282696 PMCID: PMC4969137 DOI: 10.1016/j.neuint.2016.05.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 05/03/2016] [Accepted: 05/17/2016] [Indexed: 11/21/2022]
Abstract
The cerebral cortex is organized into morphologically distinct areas that provide biological frameworks underlying perception, cognition, and behavior. Profiling mouse and human cortical transcriptomes have revealed temporal-specific differential gene expression modules in distinct neocortical areas during cortical map establishment. However, the biological roles of spatiotemporal gene expression in cortical patterning and how cortical topographic gene expression is regulated are largely unknown. Here, we characterize temporal- and spatial-defined expression of serotonin (5-HT) transporter (SERT) in glutamatergic neurons during sensory map development in mice. SERT is transiently expressed in glutamatergic thalamic neurons projecting to sensory cortices and in pyramidal neurons in the prefrontal cortex (PFC) and hippocampus (HPC) during the period that lays down the basic functional neural circuits. We previously identified that knockout of SERT in the thalamic neurons blocks 5-HT uptake by their thalamocortical axons, resulting in excessive 5-HT signaling that impairs sensory map architecture. In contrast, here we show that selective SERT knockout in the PFC and HPC neurons does not perturb sensory map patterning. These data suggest that transient SERT expression in specific glutamatergic neurons provides area-specific instructions for cortical map patterning. Hence, genetic and pharmacological manipulations of this SERT function could illuminate the fundamental genetic programming of cortex-specific maps and biological roles of temporal-specific cortical topographic gene expression in normal development and mental disorders.
Collapse
Affiliation(s)
- Xiaoning Chen
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Emilie I Petit
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kostantin Dobrenis
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA; Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ji Ying Sze
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA; Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
49
|
Sailer MHM, Sarvepalli D, Brégère C, Fisch U, Guentchev M, Weller M, Guzman R, Bettler B, Ghosh A, Hutter G. An Enzyme- and Serum-free Neural Stem Cell Culture Model for EMT Investigation Suited for Drug Discovery. J Vis Exp 2016. [PMID: 27583933 DOI: 10.3791/54018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Epithelial to mesenchymal transition (EMT) describes the process of epithelium transdifferentiating into mesenchyme. EMT is a fundamental process during embryonic development that also commonly occurs in glioblastoma, the most frequent malignant brain tumor. EMT has also been observed in multiple carcinomas outside the brain including breast cancer, lung cancer, colon cancer, gastric cancer. EMT is centrally linked to malignancy by promoting migration, invasion and metastasis formation. The mechanisms of EMT induction are not fully understood. Here we describe an in vitro system for standardized isolation of cortical neural stem cells (NSCs) and subsequent EMT-induction. This system provides the flexibility to use either single cells or explant culture. In this system, rat or mouse embryonic forebrain NSCs are cultured in a defined medium, devoid of serum and enzymes. The NSCs expressed Olig2 and Sox10, two transcription factors observed in oligodendrocyte precursor cells (OPCs). Using this system, interactions between FGF-, BMP- and TGFβ-signaling involving Zeb1, Zeb2, and Twist2 were observed where TGFβ-activation significantly enhanced cell migration, suggesting a synergistic BMP-/TGFβ-interaction. The results point to a network of FGF-, BMP- and TGFβ-signaling to be involved in EMT induction and maintenance. This model system is relevant to investigate EMT in vitro. It is cost-efficient and shows high reproducibility. It also allows for the comparison of different compounds with respect to their migration responses (quantitative distance measurement), and high-throughput screening of compounds to inhibit or enhance EMT (qualitative measurement). The model is therefore well suited to test drug libraries for substances affecting EMT.
Collapse
Affiliation(s)
| | - Durga Sarvepalli
- Molecular Signalling and Gene Therapy, Narayana Nethralaya Foundation, Narayana Health City
| | - Catherine Brégère
- Brain Ischemia and Regeneration, Department of Biomedicine, University Hospital Basel
| | - Urs Fisch
- Brain Ischemia and Regeneration, Department of Biomedicine, University Hospital Basel
| | | | - Michael Weller
- Department of Neurology, Laboratory of Molecular Neuro Oncology, University Hospital of Zurich
| | - Raphael Guzman
- Brain Ischemia and Regeneration, Department of Biomedicine, University Hospital Basel
| | | | - Arkasubhra Ghosh
- Molecular Signalling and Gene Therapy, Narayana Nethralaya Foundation, Narayana Health City
| | - Gregor Hutter
- Department of Neurosurgery and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University
| |
Collapse
|
50
|
Mason JO, Price DJ. Building brains in a dish: Prospects for growing cerebral organoids from stem cells. Neuroscience 2016; 334:105-118. [PMID: 27506142 DOI: 10.1016/j.neuroscience.2016.07.048] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 07/22/2016] [Accepted: 07/30/2016] [Indexed: 11/27/2022]
Abstract
The recent development of organoid techniques, in which embryonic brain-like tissue can be grown from human or mouse stem cells in vitro offers the potential to transform the way in which brain development is studied. In this review, we summarize key aspects of the embryonic development of mammalian forebrains, focussing in particular on the cerebral cortex and highlight significant differences between mouse and primates, including human. We discuss recent work using cerebral organoids that has revealed key similarities and differences between their development and that of the brain in vivo. Finally, we outline the ways in which cerebral organoids can be used in combination with CRISPR/Cas9 genome editing to unravel genetic mechanisms that control embryonic development of the cerebral cortex, how this can help us understand the causes of neurodevelopmental disorders and some of the key challenges which will have to be resolved before organoids can become a mainstream tool to study brain development.
Collapse
Affiliation(s)
- John O Mason
- Centre for Integrative Physiology, Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK.
| | - David J Price
- Centre for Integrative Physiology, Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK.
| |
Collapse
|