1
|
Wu B, Li J, Jin X. Every cell every gene all at once: Systems genetic approaches toward corticogenesis. Curr Opin Neurobiol 2025; 92:103034. [PMID: 40339387 DOI: 10.1016/j.conb.2025.103034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 12/24/2024] [Accepted: 04/09/2025] [Indexed: 05/10/2025]
Abstract
The development of the cerebral cortex is a stepwise process that involves numerous cell types and signaling pathways to achieve the functional assembly of neural circuits. Our understanding of this process is primarily rooted in findings from studying transgenic knockout models, which reveal coordinated molecular actions, particularly transcription factor cascades critical for cell type acquisition and maintenance in a context-dependent manner. Further resolving their cell type specificity necessitates the use of high-throughput, high-content methodologies. Over the past decade, the emerging single-cell genomics and in vivo CRISPR tools have provided new approaches to study neurodevelopment with elevated scale and resolution. In this review, we discussed efforts to study mouse cortical cell fate determination using single-cell genomics methods. Additionally, we explored recent studies combining programmable gene editing with single-cell phenotypic assays to investigate the function of transcription factors in perinatal cortical development, delineating cell-type specific, functional cytoarchitecture of the developing brain.
Collapse
Affiliation(s)
- Boli Wu
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jiwen Li
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Xin Jin
- Department of Neuroscience, Dorris Neuroscience Center, Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
2
|
Konstantinides N, Desplan C. Neuronal Circuit Evolution: From Development to Structure and Adaptive Significance. Cold Spring Harb Perspect Biol 2025; 17:a041493. [PMID: 38951021 PMCID: PMC11688512 DOI: 10.1101/cshperspect.a041493] [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: 07/03/2024]
Abstract
Neuronal circuits represent the functional units of the brain. Understanding how the circuits are generated to perform computations will help us understand how the brain functions. Nevertheless, neuronal circuits are not engineered, but have formed through millions of years of animal evolution. We posit that it is necessary to study neuronal circuit evolution to comprehensively understand circuit function. Here, we review our current knowledge regarding the mechanisms that underlie circuit evolution. First, we describe the possible genetic and developmental mechanisms that have contributed to circuit evolution. Then, we discuss the structural changes of circuits during evolution and how these changes affected circuit function. Finally, we try to put circuit evolution in an ecological context and assess the adaptive significance of specific examples. We argue that, thanks to the advent of new tools and technologies, evolutionary neurobiology now allows us to address questions regarding the evolution of circuitry and behavior that were unimaginable until very recently.
Collapse
Affiliation(s)
| | - Claude Desplan
- Department of Biology, New York University, New York, New York 10003, USA
| |
Collapse
|
3
|
He Z, Treutlein B. Gene programs driving cortical neuron specifications. Nat Neurosci 2025; 28:919-920. [PMID: 40259072 DOI: 10.1038/s41593-025-01934-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Affiliation(s)
- Zhisong He
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland.
| | - Barbara Treutlein
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland.
| |
Collapse
|
4
|
Nano PR, Fazzari E, Azizad D, Martija A, Nguyen CV, Wang S, Giang V, Kan RL, Yoo J, Wick B, Haeussler M, Bhaduri A. Integrated analysis of molecular atlases unveils modules driving developmental cell subtype specification in the human cortex. Nat Neurosci 2025; 28:949-963. [PMID: 40259073 PMCID: PMC12081304 DOI: 10.1038/s41593-025-01933-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 02/27/2025] [Indexed: 04/23/2025]
Abstract
Human brain development requires generating diverse cell types, a process explored by single-cell transcriptomics. Through parallel meta-analyses of the human cortex in development (seven datasets) and adulthood (16 datasets), we generated over 500 gene co-expression networks that can describe mechanisms of cortical development, centering on peak stages of neurogenesis. These meta-modules show dynamic cell subtype specificities throughout cortical development, with several developmental meta-modules displaying spatiotemporal expression patterns that allude to potential roles in cell fate specification. We validated the expression of these modules in primary human cortical tissues. These include meta-module 20, a module elevated in FEZF2+ deep layer neurons that includes TSHZ3, a transcription factor associated with neurodevelopmental disorders. Human cortical chimeroid experiments validated that both FEZF2 and TSHZ3 are required to drive module 20 activity and deep layer neuron specification but through distinct modalities. These studies demonstrate how meta-atlases can engender further mechanistic analyses of cortical fate specification.
Collapse
Grants
- UM1 MH130991 NIMH NIH HHS
- T32 NS048004 NINDS NIH HHS
- R01MH132689 U.S. Department of Health & Human Services | NIH | National Institute of Mental Health (NIMH)
- RF1 MH132662 NIMH NIH HHS
- T32 GM008243 NIGMS NIH HHS
- R00 NS111731 NINDS NIH HHS
- R00NS111731 U.S. Department of Health & Human Services | NIH | National Institute of Neurological Disorders and Stroke (NINDS)
- R01 MH132689 NIMH NIH HHS
- T32 GM145388 NIGMS NIH HHS
- U24 HG002371 NHGRI NIH HHS
- U.S. Department of Health & Human Services | NIH | National Institute of Mental Health (NIMH)
- U.S. Department of Health & Human Services | NIH | National Institute of Neurological Disorders and Stroke (NINDS)
- We would like to thank the members of the Bhaduri Lab for their insightful advice and comments on the study. We would like to thank the Broad Stem Cell Research Center Flow Cytometry core for their help in isolating cells for this project, Charina Julian for help with running sequencing, and Dr. Laurent Fasano for generously sharing the antibody against TSHZ3. The work performed in the manuscript was generously funded by R00NS111731 from the NIH (NINDS), R01MH132689 from the NIH (NIMH), the Young Investigator Award from the Brain & Behavior Research Foundation, the Alfred P. Sloan Foundation, the Rose Hills Foundation, and the Klingenstein-Simons Fellowship from the Esther A. & Joseph Klingenstein Fund and the Simons Foundation (to A.B.). Additional funding was provided to P.R.N. (UCLA Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research Training Program, UCLA Intercampus Medical Genetics Training Program (USHHS Ruth L. Kirschstein Institutional National Research Service Award # T32GM008243)), C.V.N. (T32 NS048004, Predoctoral Fellowship in association with the Training Grant in Neurobehavioral Genetics), and R.K. (T32 GM145388, Cell and Molecular Biology Training Program), and M.H. (NIMH BRAIN NIMH RF1MH132662, NHGRI U24HG002371, CIRM DISC0-14514 (with A.B.)).
Collapse
Affiliation(s)
- Patricia R Nano
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Elisa Fazzari
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Daria Azizad
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Antoni Martija
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Claudia V Nguyen
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sean Wang
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Vanna Giang
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ryan L Kan
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Juyoun Yoo
- Department of Neuroscience, Columbia University, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Brittney Wick
- Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA, USA
| | | | - Aparna Bhaduri
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
5
|
Tobias IC, Moorthy SD, Shchuka VM, Langroudi L, Cherednychenko M, Gillespie ZE, Duncan AG, Tian R, Gajewska NA, Di Roberto RB, Mitchell JA. A Sox2 enhancer cluster regulates region-specific neural fates from mouse embryonic stem cells. G3 (BETHESDA, MD.) 2025; 15:jkaf012. [PMID: 39849901 PMCID: PMC12005160 DOI: 10.1093/g3journal/jkaf012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/14/2025] [Accepted: 01/19/2025] [Indexed: 01/25/2025]
Abstract
Sex-determining region Y box 2 (Sox2) is a critical transcription factor for embryogenesis and neural stem and progenitor cell (NSPC) maintenance. While distal enhancers control Sox2 in embryonic stem cells (ESCs), enhancers closer to the gene are implicated in Sox2 transcriptional regulation in neural development. We hypothesize that a downstream enhancer cluster, termed Sox2 regulatory regions 2-18 (SRR2-18), regulates Sox2 transcription in neural stem cells and we investigate this in NSPCs derived from mouse ESCs. Using functional genomics and CRISPR-Cas9-mediated deletion analyses, we investigate the role of SRR2-18 in Sox2 regulation during neural differentiation. Transcriptome analyses demonstrate that the loss of even 1 copy of SRR2-18 disrupts the region-specific identity of NSPCs, reducing the expression of genes associated with more anterior regions of the embryonic nervous system. Homozygous deletion of this Sox2 neural enhancer cluster causes reduced SOX2 protein, less frequent interaction with transcriptional machinery, and leads to perturbed chromatin accessibility genome-wide further affecting the expression of neurodevelopmental and anterior-posterior regionalization genes. Furthermore, homozygous NSPC deletants exhibit self-renewal defects and impaired differentiation into cell types found in the brain. Altogether, our data define a cis-regulatory enhancer cluster controlling Sox2 transcription in NSPCs and highlight the sensitivity of neural differentiation processes to decreased Sox2 transcription, which causes differentiation into posterior neural fates, specifically the caudal neural tube. This study highlights the importance of precise Sox2 regulation by SRR2-18 in neural differentiation.
Collapse
Affiliation(s)
- Ian C Tobias
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Sakthi D Moorthy
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Virlana M Shchuka
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Lida Langroudi
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Mariia Cherednychenko
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Zoe E Gillespie
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Andrew G Duncan
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Ruxiao Tian
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Natalia A Gajewska
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Raphaël B Di Roberto
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| | - Jennifer A Mitchell
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario M5S 3G5, Canada
| |
Collapse
|
6
|
Hanson MA, Bibi N, Safa A, Nagarajan D, Marshall AH, Johantges AC, Wester JC. Development of Differential Sublaminar Feedforward Inhibitory Circuits in CA1 Hippocampus Requires Satb2. J Neurosci 2025; 45:e0737242024. [PMID: 39753301 PMCID: PMC11841754 DOI: 10.1523/jneurosci.0737-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 10/30/2024] [Accepted: 12/04/2024] [Indexed: 01/07/2025] Open
Abstract
Pyramidal cells (PCs) in CA1 hippocampus can be classified by their radial position as deep or superficial and organize into subtype-specific circuits necessary for differential information processing. Specifically, superficial PCs receive fewer inhibitory synapses from parvalbumin (PV)-expressing interneurons than deep PCs, resulting in weaker feedforward inhibition of input from CA3 Schaffer collaterals. Using mice, we investigated mechanisms underlying CA1 PC differentiation and the development of this inhibitory circuit motif. We found that the transcriptional regulator SATB2, which is necessary for pyramidal cell differentiation in the neocortex, is selectively expressed in superficial PCs during early postnatal development. To investigate its role in CA1, we conditionally knocked out Satb2 from pyramidal cells during embryonic development using both male and female Emx1IRES-Cre; Satb2flox/flox mice. Loss of Satb2 resulted in increased feedforward inhibition of CA3 Schaffer collateral input to superficial PCs, which matched that observed to deep PCs in control mice. Using paired whole-cell recordings between PCs and PV+ interneurons, we found this was due to an increase in the strength of unitary inhibitory synaptic connections from PV+ interneurons to mutant superficial PCs. Regulation of synapse strength was restricted to inhibitory synapses; excitatory synaptic connections from CA3 to CA1 PCs and CA1 PCs to PV+ interneurons were not affected by loss of Satb2 Finally, we show that SATB2 expression in superficial PCs is necessary to suppress the formation of synapses from PV+ interneurons during synaptogenesis. Thus, early postnatal expression of SATB2 in superficial PCs is necessary for the development of biased feedforward inhibition in CA1.
Collapse
Affiliation(s)
- Meretta A Hanson
- Department of Neuroscience, The Ohio State University College of Medicine, Columbus, Ohio 43210
- Neuroscience Graduate Program, The Ohio State University, Columbus, Ohio 43210
| | - Noor Bibi
- Department of Neuroscience, The Ohio State University College of Medicine, Columbus, Ohio 43210
| | - Alireza Safa
- Department of Neuroscience, The Ohio State University College of Medicine, Columbus, Ohio 43210
| | - Devipriyanka Nagarajan
- Department of Neuroscience, The Ohio State University College of Medicine, Columbus, Ohio 43210
| | - Alec H Marshall
- Department of Neuroscience, The Ohio State University College of Medicine, Columbus, Ohio 43210
- Neuroscience Graduate Program, The Ohio State University, Columbus, Ohio 43210
| | - Aidan C Johantges
- Department of Neuroscience, The Ohio State University College of Medicine, Columbus, Ohio 43210
| | - Jason C Wester
- Department of Neuroscience, The Ohio State University College of Medicine, Columbus, Ohio 43210
| |
Collapse
|
7
|
Passos GA, Genari AB, Assis AF, Monteleone-Cassiano AC, Donadi EA, Oliveira EH, Duarte MJ, Machado MV, Tanaka PP, Mascarenhas R. The Thymus as a Mirror of the Body's Gene Expression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1471:247-268. [PMID: 40067590 DOI: 10.1007/978-3-031-77921-3_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
The thymus, a complex organ formed by different cell types that establish close interaction, serves a unique function of significant interest. The role played by the thymic stroma is not only a connective tissue or a support structure, but it also involves the stromal thymic epithelial cells (TECs) establishing physical and functional interaction with developing thymocytes. This interaction culminates in the induction of central tolerance, a function that sets this organ apart. The role played by the medullary thymic epithelial cells (mTECs) is noteworthy and is the focus of many studies. The transcriptome of mTEC cells is also very complex. These cells express nearly the functional genome without altering morphological and functional features. Among the thousand mRNAs expressed, a particular set encodes all peripheral tissue antigens (PTAs), representing the body's different tissues and organs. The consequence of ectopic proteins translated from these mRNAs in the thymus is immunological and is associated with self-nonself-discrimination and induction of central tolerance. Due to the wide variety of PTAs, this process was termed promiscuous gene expression (PGE), whose control is shared between autoimmune regulator (human AIRE/murine Aire), a transcriptional modulator, and forebrain-expressed zinc finger 2 (FEZF2/Fezf2), a transcription factor. Therefore, this molecular-genetic process is closely linked to eliminating autoreactive thymocytes in the thymus through negative selection. In this chapter, we review PGE in mTECs and its immunologic implication, the role of the Aire and Fezf2genes, the role of Aire on the expression of miRNAs in mTECs, its consequence on PGE and the manipulation of the Aire expression either by siRNA or by genome editing using the Crispr-Cas9 system.
Collapse
Affiliation(s)
- Geraldo A Passos
- Laboratory of Genetics and Molecular Biology, Department of Basic and Oral Biology, Ribeirão Preto School of Dentistry (FORP-USP), University of São Paulo, Ribeirão Preto, SP, Brazil.
| | - Adriana B Genari
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Amanda F Assis
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Ana C Monteleone-Cassiano
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Eduardo A Donadi
- Department of Clinical Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Ernna H Oliveira
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Max J Duarte
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Mayara V Machado
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Pedro P Tanaka
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Romário Mascarenhas
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
8
|
Iyer A, Vaasjo LO, Siththanandan VB, K C R, Thurmon A, Akumuo M, Lu V, Nnebe C, Nair R, Galazo MJ, Tharin S. miR-193b-365 microcluster downstream of Fezf2 coordinates neuron-subtype identity and dendritic morphology in cortical projection neurons. iScience 2024; 27:111500. [PMID: 39759000 PMCID: PMC11697703 DOI: 10.1016/j.isci.2024.111500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 07/30/2024] [Accepted: 11/26/2024] [Indexed: 01/07/2025] Open
Abstract
Different neuron types develop characteristic axonal and dendritic arborizations that determine their inputs, outputs, and functions. Expression of fate-determinant transcription factors is essential for specification of their distinct identities. However, the mechanisms downstream of fate-determinant factors coordinating different aspects of neuron identity are not understood. Specifically, how distinct projection neurons develop appropriate dendritic arbors that determine their inputs is unknown. Here, we investigate this question in corticospinal and callosal projection neurons. We identified a mechanism linking the corticospinal/corticofugal identity gene Fezf2 with the regulation of dendritic development. We show that miR-193b∼365 microRNA cluster is regulated by Fezf2 and enriched in corticospinal neurons. miR-193b∼365 represses mitogen-activated protein kinase 8 (MAPK8) to regulate corticospinal dendritic development. miR-193b∼365 overexpression in callosal neurons abnormally reduces MAPK8 signal and dendritic complexity. Our findings show that regulation of MAPK8 via miR-193b∼365 cluster regulates dendritic development, providing a mechanism that coordinates projection neuron identity, specified by Fezf2, and neuron-specific dendritic morphology.
Collapse
Affiliation(s)
- Asha Iyer
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Lee O. Vaasjo
- Neuroscience program, Tulane Brain Institute, Tulane University, New Orleans, LA 70118 USA
| | | | - Rajan K C
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118 USA
| | - Abbigail Thurmon
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118 USA
| | - Mauren Akumuo
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118 USA
| | - Victoria Lu
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
| | - Chelsea Nnebe
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
- Neurosciences PhD program, Stanford University, Stanford, CA 94305, USA
| | - Ramesh Nair
- Stanford Center for Genomics and Personalized Medicine, Stanford, CA 94305, USA
| | - Maria J. Galazo
- Neuroscience program, Tulane Brain Institute, Tulane University, New Orleans, LA 70118 USA
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118 USA
| | - Suzanne Tharin
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA
- Division of Neurosurgery, Palo Alto Veterans Affairs Health Care System, Palo Alto, CA 94304, USA
- Neurosciences PhD program, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
9
|
Wu R, Ramakrishnan S, Lin H, Dong Z, Liu M, Qiang L. Development and validation a novel FEZF2 based fluorescent reporter for corticospinal motor neurons. Metab Brain Dis 2024; 40:17. [PMID: 39560839 PMCID: PMC11654391 DOI: 10.1007/s11011-024-01482-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 11/12/2024] [Indexed: 11/20/2024]
Abstract
Corticospinal motor neurons (CSMNs), also named upper motor neurons, are the giant pyramidal neurons called Betz cells. In mammals, the majority of CSMNs reside within layer V of the primary motor cortex, where they extend long axon bundles named the pyramidal tract into the brainstem and the spinal cord to control voluntary movement. CSMN lesions are implicated in a variety of neurodegenerative disorders, such Amyotrophic Lateral Sclerosis, Primary Lateral Sclerosis and Hereditary Spastic paraplegia. Although FEZF2-CTIP2 genetic axis have been indicated as the cardinal molecular pathway underlying the development of CSMNs, these proteins are transcription factors that are mostly used to label the nuclei of CSMNs in the fixed cells and tissues. Therefore, a fluorescent reporter to mark CSMNs will be invaluable in identifying living CSMNs, including their extended processes, for time-lapse imaging and high-throughput molecular analyses with much more improved specificity. Based on the in-silico analysis, we identified a putative region within the promoter sequence of FEZF2 and assembled it with an indispensable enhancer motif at its downstream of the gene to form a complex promoter that drives the expression of reporter GFP. The plasmid and virus of FEZF2:eGFP reporter constructs were further validated for its use in specifically labeling CSMNs in primary neuronal cultures from the embryonic rat motor cortex, postnatal mouse cortex. This innovative molecular labeling tool has the potential to offer indispensable support in diverse experimental setups, enabling a comprehensive understanding of the susceptibility and specificity of CSMNs in a wide array of neurological disorders.
Collapse
Affiliation(s)
- Ronghua Wu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Skandha Ramakrishnan
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA, 19129, USA
| | - Haixu Lin
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Zhangji Dong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China
| | - Mei Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu, China.
| | - Liang Qiang
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA, 19129, USA.
| |
Collapse
|
10
|
Abe P, Lavalley A, Morassut I, Santinha AJ, Roig-Puiggros S, Javed A, Klingler E, Baumann N, Prados J, Platt RJ, Jabaudon D. Molecular programs guiding arealization of descending cortical pathways. Nature 2024; 634:644-651. [PMID: 39261725 DOI: 10.1038/s41586-024-07895-y] [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: 07/13/2023] [Accepted: 08/01/2024] [Indexed: 09/13/2024]
Abstract
Layer 5 extratelencephalic (ET) neurons are present across neocortical areas and send axons to multiple subcortical targets1-6. Two cardinal subtypes exist7,8: (1) Slco2a1-expressing neurons (ETdist), which predominate in the motor cortex and project distally to the pons, medulla and spinal cord; and (2) Nprs1- or Hpgd-expressing neurons (ETprox), which predominate in the visual cortex and project more proximally to the pons and thalamus. An understanding of how area-specific ETdist and ETprox emerge during development is important because they are critical for fine motor skills and are susceptible to spinal cord injury and amyotrophic lateral sclerosis9-12. Here, using cross-areal mapping of axonal projections in the mouse neocortex, we identify the subtype-specific developmental dynamics of ET neurons. Whereas subsets of ETprox emerge by pruning of ETdist axons, others emerge de novo. We outline corresponding subtype-specific developmental transcriptional programs using single-nucleus sequencing. Leveraging these findings, we use postnatal in vivo knockdown of subtype-specific transcription factors to reprogram ET neuron connectivity towards more proximal targets. Together, these results show the functional transcriptional programs driving ET neuron diversity and uncover cell subtype-specific gene regulatory networks that can be manipulated to direct target specificity in motor corticofugal pathways.
Collapse
Affiliation(s)
- Philipp Abe
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Dresden, Germany
| | - Adrien Lavalley
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
- Department of Clinical Neurosciences, Geneva University Hospital, Geneva, Switzerland
| | - Ilaria Morassut
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Antonio J Santinha
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Sergi Roig-Puiggros
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Awais Javed
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Esther Klingler
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
| | - Natalia Baumann
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Julien Prados
- Bioinformatic Support Platform, University of Geneva, Geneva, Switzerland
| | - Randall J Platt
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- Basel Research Center for Child Health, Basel, Switzerland
- Department of Chemistry, University of Basel, Basel, Switzerland
- NCCR Molecular Systems Engineering, Basel, Switzerland
| | - Denis Jabaudon
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland.
- Department of Clinical Neurosciences, Geneva University Hospital, Geneva, Switzerland.
- Université Paris Cité, Imagine Institute, Paris, France.
| |
Collapse
|
11
|
Di Bella DJ, Domínguez-Iturza N, Brown JR, Arlotta P. Making Ramón y Cajal proud: Development of cell identity and diversity in the cerebral cortex. Neuron 2024; 112:2091-2111. [PMID: 38754415 PMCID: PMC11771131 DOI: 10.1016/j.neuron.2024.04.021] [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: 12/15/2023] [Revised: 03/28/2024] [Accepted: 04/18/2024] [Indexed: 05/18/2024]
Abstract
Since the beautiful images of Santiago Ramón y Cajal provided a first glimpse into the immense diversity and complexity of cell types found in the cerebral cortex, neuroscience has been challenged and inspired to understand how these diverse cells are generated and how they interact with each other to orchestrate the development of this remarkable tissue. Some fundamental questions drive the field's quest to understand cortical development: what are the mechanistic principles that govern the emergence of neuronal diversity? How do extrinsic and intrinsic signals integrate with physical forces and activity to shape cell identity? How do the diverse populations of neurons and glia influence each other during development to guarantee proper integration and function? The advent of powerful new technologies to profile and perturb cortical development at unprecedented resolution and across a variety of modalities has offered a new opportunity to integrate past knowledge with brand new data. Here, we review some of this progress using cortical excitatory projection neurons as a system to draw out general principles of cell diversification and the role of cell-cell interactions during cortical development.
Collapse
Affiliation(s)
- Daniela J Di Bella
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Nuria Domínguez-Iturza
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Juliana R Brown
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Paola Arlotta
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
12
|
Simon CM, Delestree N, Montes J, Gerstner F, Carranza E, Sowoidnich L, Buettner JM, Pagiazitis JG, Prat-Ortega G, Ensel S, Donadio S, Garcia JL, Kratimenos P, Chung WK, Sumner CJ, Weimer LH, Pirondini E, Capogrosso M, Pellizzoni L, De Vivo DC, Mentis GZ. Dysfunction of proprioceptive sensory synapses is a pathogenic event and therapeutic target in mice and humans with spinal muscular atrophy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.03.24308132. [PMID: 38883729 PMCID: PMC11177917 DOI: 10.1101/2024.06.03.24308132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Spinal muscular atrophy (SMA) is a neurodegenerative disease characterized by a varying degree of severity that correlates with the reduction of SMN protein levels. Motor neuron degeneration and skeletal muscle atrophy are hallmarks of SMA, but it is unknown whether other mechanisms contribute to the spectrum of clinical phenotypes. Here, through a combination of physiological and morphological studies in mouse models and SMA patients, we identify dysfunction and loss of proprioceptive sensory synapses as key signatures of SMA pathology. We demonstrate that SMA patients exhibit impaired proprioception, and their proprioceptive sensory synapses are dysfunctional as measured by the neurophysiological test of the Hoffmann reflex (H-reflex). We further show that loss of excitatory afferent synapses and altered potassium channel expression in SMA motor neurons are conserved pathogenic events found in both severely affected patients and mouse models. Lastly, we report that improved motor function and fatigability in ambulatory SMA patients and mouse models treated with SMN-inducing drugs correlate with increased function of sensory-motor circuits that can be accurately captured by the H-reflex assay. Thus, sensory synaptic dysfunction is a clinically relevant event in SMA, and the H-reflex is a suitable assay to monitor disease progression and treatment efficacy of motor circuit pathology.
Collapse
Affiliation(s)
- CM Simon
- Center for Motor Neuron Biology and Disease, Columbia University, NY, USA
- Dept. of Neurology, Columbia University, NY, USA
- Carl-Ludwig-Institute for Physiology, Leipzig University, Leipzig, Germany
| | - N Delestree
- Center for Motor Neuron Biology and Disease, Columbia University, NY, USA
- Dept. of Neurology, Columbia University, NY, USA
| | - J Montes
- Center for Motor Neuron Biology and Disease, Columbia University, NY, USA
- Dept. of Rehabilitation and Regenerative Medicine, Columbia University, NY, USA
| | - F Gerstner
- Carl-Ludwig-Institute for Physiology, Leipzig University, Leipzig, Germany
| | - E Carranza
- Depts. Physical Medicine & Rehabilitation & Bioengineering, University of Pittsburgh, PA, USA
- Rehab and Neural Engineering Labs, University of Pittsburgh, PA, USA
| | - L Sowoidnich
- Carl-Ludwig-Institute for Physiology, Leipzig University, Leipzig, Germany
| | - JM Buettner
- Carl-Ludwig-Institute for Physiology, Leipzig University, Leipzig, Germany
| | - JG Pagiazitis
- Center for Motor Neuron Biology and Disease, Columbia University, NY, USA
- Dept. of Neurology, Columbia University, NY, USA
| | - G Prat-Ortega
- Rehab and Neural Engineering Labs, University of Pittsburgh, PA, USA
- Depts. of Neurological Surgery & Bioengineering, University of Pittsburgh, PA, USA
| | - S Ensel
- Rehab and Neural Engineering Labs, University of Pittsburgh, PA, USA
- Depts. of Neurological Surgery & Bioengineering, University of Pittsburgh, PA, USA
| | - S Donadio
- Rehab and Neural Engineering Labs, University of Pittsburgh, PA, USA
- Depts. of Neurological Surgery & Bioengineering, University of Pittsburgh, PA, USA
| | - JL Garcia
- Dept. of Neurology, Columbia University, NY, USA
| | - P Kratimenos
- Center for Neuroscience Research, Children’s National Res. Institute, Washington, DC, USA
- Dept. of Pediatrics, G Washington Univ. Sch. of Medicine, Washington, DC, USA
| | - WK Chung
- Dept. of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA USA
| | - CJ Sumner
- Depts. of Neurology, Neuroscience and Genetic Medicine, Johns Hopkins University School of Medicine, MD, USA
| | - LH Weimer
- Dept. of Neurology, Columbia University, NY, USA
| | - E Pirondini
- Depts. Physical Medicine & Rehabilitation & Bioengineering, University of Pittsburgh, PA, USA
- Rehab and Neural Engineering Labs, University of Pittsburgh, PA, USA
| | - M Capogrosso
- Rehab and Neural Engineering Labs, University of Pittsburgh, PA, USA
- Depts. of Neurological Surgery & Bioengineering, University of Pittsburgh, PA, USA
| | - L Pellizzoni
- Center for Motor Neuron Biology and Disease, Columbia University, NY, USA
- Dept. of Neurology, Columbia University, NY, USA
- Dept. of Pathology and Cell Biology, Columbia University, NY, USA
| | - DC De Vivo
- Center for Motor Neuron Biology and Disease, Columbia University, NY, USA
- Dept. of Neurology, Columbia University, NY, USA
| | - GZ Mentis
- Center for Motor Neuron Biology and Disease, Columbia University, NY, USA
- Dept. of Neurology, Columbia University, NY, USA
- Dept. of Pathology and Cell Biology, Columbia University, NY, USA
| |
Collapse
|
13
|
Ozkan A, Padmanabhan HK, Shipman SL, Azim E, Kumar P, Sadegh C, Basak AN, Macklis JD. Directed differentiation of functional corticospinal-like neurons from endogenous SOX6+/NG2+ cortical progenitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.21.590488. [PMID: 38712174 PMCID: PMC11071355 DOI: 10.1101/2024.04.21.590488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Corticospinal neurons (CSN) centrally degenerate in amyotrophic lateral sclerosis (ALS), along with spinal motor neurons, and loss of voluntary motor function in spinal cord injury (SCI) results from damage to CSN axons. For functional regeneration of specifically affected neuronal circuitry in vivo , or for optimally informative disease modeling and/or therapeutic screening in vitro , it is important to reproduce the type or subtype of neurons involved. No such appropriate in vitro models exist with which to investigate CSN selective vulnerability and degeneration in ALS, or to investigate routes to regeneration of CSN circuitry for ALS or SCI, critically limiting the relevance of much research. Here, we identify that the HMG-domain transcription factor Sox6 is expressed by a subset of NG2+ endogenous cortical progenitors in postnatal and adult cortex, and that Sox6 suppresses a latent neurogenic program by repressing inappropriate proneural Neurog2 expression by progenitors. We FACS-purify these genetically accessible progenitors from postnatal mouse cortex and establish a pure culture system to investigate their potential for directed differentiation into CSN. We then employ a multi-component construct with complementary and differentiation-sharpening transcriptional controls (activating Neurog2, Fezf2 , while antagonizing Olig2 with VP16:Olig2 ). We generate corticospinal-like neurons from SOX6+/NG2+ cortical progenitors, and find that these neurons differentiate with remarkable fidelity compared with corticospinal neurons in vivo . They possess appropriate morphological, molecular, transcriptomic, and electrophysiological characteristics, without characteristics of the alternate intracortical or other neuronal subtypes. We identify that these critical specifics of differentiation are not reproduced by commonly employed Neurog2 -driven differentiation. Neurons induced by Neurog2 instead exhibit aberrant multi-axon morphology and express molecular hallmarks of alternate cortical projection subtypes, often in mixed form. Together, this developmentally-based directed differentiation from genetically accessible cortical progenitors sets a precedent and foundation for in vitro mechanistic and therapeutic disease modeling, and toward regenerative neuronal repopulation and circuit repair.
Collapse
|
14
|
Assali A, Chenaux G, Cho JY, Berto S, Ehrlich NA, Cowan CW. EphB1 controls long-range cortical axon guidance through a cell non-autonomous role in GABAergic cells. Development 2024; 151:dev201439. [PMID: 38345254 PMCID: PMC10946438 DOI: 10.1242/dev.201439] [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: 11/15/2022] [Accepted: 01/28/2024] [Indexed: 02/29/2024]
Abstract
EphB1 is required for proper guidance of cortical axon projections during brain development, but how EphB1 regulates this process remains unclear. We show here that EphB1 conditional knockout (cKO) in GABAergic cells (Vgat-Cre), but not in cortical excitatory neurons (Emx1-Cre), reproduced the cortical axon guidance defects observed in global EphB1 KO mice. Interestingly, in EphB1 cKOVgat mice, the misguided axon bundles contained co-mingled striatal GABAergic and somatosensory cortical glutamatergic axons. In wild-type mice, somatosensory axons also co-fasciculated with striatal axons, notably in the globus pallidus, suggesting that a subset of glutamatergic cortical axons normally follows long-range GABAergic axons to reach their targets. Surprisingly, the ectopic axons in EphB1 KO mice were juxtaposed to major blood vessels. However, conditional loss of EphB1 in endothelial cells (Tie2-Cre) did not produce the axon guidance defects, suggesting that EphB1 in GABAergic neurons normally promotes avoidance of these ectopic axons from the developing brain vasculature. Together, our data reveal a new role for EphB1 in GABAergic neurons to influence proper cortical glutamatergic axon guidance during brain development.
Collapse
Affiliation(s)
- Ahlem Assali
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - George Chenaux
- Department of Psychiatry, University of Texas Southwestern Medical School, Dallas, TX 75390, USA
| | - Jennifer Y. Cho
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Stefano Berto
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Nathan A. Ehrlich
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Christopher W. Cowan
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29425, USA
- Department of Psychiatry, University of Texas Southwestern Medical School, Dallas, TX 75390, USA
| |
Collapse
|
15
|
Bakalar D, Gavrilova O, Jiang SZ, Zhang HY, Roy S, Williams SK, Liu N, Wisser S, Usdin TB, Eiden LE. Constitutive and conditional deletion reveals distinct phenotypes driven by developmental versus neurotransmitter actions of the neuropeptide PACAP. J Neuroendocrinol 2023; 35:e13286. [PMID: 37309259 PMCID: PMC10620107 DOI: 10.1111/jne.13286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/11/2023] [Accepted: 04/25/2023] [Indexed: 06/14/2023]
Abstract
Neuropeptides may exert trophic effects during development, and then neurotransmitter roles in the developed nervous system. One way to associate peptide-deficiency phenotypes with either role is first to assess potential phenotypes in so-called constitutive knockout mice, and then proceed to specify, regionally and temporally, where and when neuropeptide expression is required to prevent these phenotypes. We have previously demonstrated that the well-known constellation of behavioral and metabolic phenotypes associated with constitutive pituitary adenylate cyclase-activating peptide (PACAP) knockout mice are accompanied by transcriptomic alterations of two types: those that distinguish the PACAP-null phenotype from wild-type (WT) in otherwise quiescent mice (cPRGs), and gene induction that occurs in response to acute environmental perturbation in WT mice that do not occur in knockout mice (aPRGs). Comparing constitutive PACAP knockout mice to a variety of temporally and regionally specific PACAP knockouts, we show that the prominent hyperlocomotor phenotype is a consequence of early loss of PACAP expression, is associated with Fos overexpression in hippocampus and basal ganglia, and that a thermoregulatory effect previously shown to be mediated by PACAP-expressing neurons of medial preoptic hypothalamus is independent of PACAP expression in those neurons in adult mice. In contrast, PACAP dependence of weight loss/hypophagia triggered by restraint stress, seen in constitutive PACAP knockout mice, is phenocopied in mice in which PACAP is deleted after neuronal differentiation. Our results imply that PACAP has a prominent role as a trophic factor early in development determining global central nervous system characteristics, and in addition a second, discrete set of functions as a neurotransmitter in the fully developed nervous system that support physiological and psychological responses to stress.
Collapse
Affiliation(s)
- Dana Bakalar
- Section on Molecular Neuroscience, National Institute of Mental Heath - Intramural Research Program, Bethesda, Maryland, USA
| | - Oksana Gavrilova
- Mouse Metabolism Core Laboratory, National Institute of Diabetes and Kidney Disease- Intramural Research Program, Bethesda, Maryland, USA
| | - Sunny Z Jiang
- Section on Molecular Neuroscience, National Institute of Mental Heath - Intramural Research Program, Bethesda, Maryland, USA
| | - Hai-Ying Zhang
- Section on Molecular Neuroscience, National Institute of Mental Heath - Intramural Research Program, Bethesda, Maryland, USA
| | - Snehashis Roy
- Systems Neuroscience Imaging Resource, National Institute of Mental Heath - Intramural Research Program, Bethesda, Maryland, USA
| | - Sarah K Williams
- Systems Neuroscience Imaging Resource, National Institute of Mental Heath - Intramural Research Program, Bethesda, Maryland, USA
| | - Naili Liu
- Mouse Metabolism Core Laboratory, National Institute of Diabetes and Kidney Disease- Intramural Research Program, Bethesda, Maryland, USA
| | - Stephen Wisser
- Systems Neuroscience Imaging Resource, National Institute of Mental Heath - Intramural Research Program, Bethesda, Maryland, USA
| | - Ted B Usdin
- Systems Neuroscience Imaging Resource, National Institute of Mental Heath - Intramural Research Program, Bethesda, Maryland, USA
| | - Lee E Eiden
- Section on Molecular Neuroscience, National Institute of Mental Heath - Intramural Research Program, Bethesda, Maryland, USA
| |
Collapse
|
16
|
Nano PR, Fazzari E, Azizad D, Nguyen CV, Wang S, Kan RL, Wick B, Haeussler M, Bhaduri A. A Meta-Atlas of the Developing Human Cortex Identifies Modules Driving Cell Subtype Specification. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.12.557406. [PMID: 37745597 PMCID: PMC10515829 DOI: 10.1101/2023.09.12.557406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Human brain development requires the generation of hundreds of diverse cell types, a process targeted by recent single-cell transcriptomic profiling efforts. Through a meta-analysis of seven of these published datasets, we have generated 225 meta-modules - gene co-expression networks that can describe mechanisms underlying cortical development. Several meta-modules have potential roles in both establishing and refining cortical cell type identities, and we validated their spatiotemporal expression in primary human cortical tissues. These include meta-module 20, associated with FEZF2+ deep layer neurons. Half of meta-module 20 genes are putative FEZF2 targets, including TSHZ3, a transcription factor associated with neurodevelopmental disorders. Human cortical organoid experiments validated that both factors are necessary for deep layer neuron specification. Importantly, subtle manipulations of these factors drive slight changes in meta-module activity that cascade into strong differences in cell fate - demonstrating how of our meta-atlas can engender further mechanistic analyses of cortical fate specification.
Collapse
|
17
|
Galazo MJ, Sweetser DA, Macklis JD. Tle4 controls both developmental acquisition and early post-natal maturation of corticothalamic projection neuron identity. Cell Rep 2023; 42:112957. [PMID: 37561632 PMCID: PMC10542749 DOI: 10.1016/j.celrep.2023.112957] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 04/21/2023] [Accepted: 07/25/2023] [Indexed: 08/12/2023] Open
Abstract
Identities of distinct neuron subtypes are specified during embryonic development, then maintained during post-natal maturation. In cerebral cortex, mechanisms controlling early acquisition of neuron-subtype identities have become increasingly understood. However, mechanisms controlling neuron-subtype identity stability during post-natal maturation are largely unexplored. We identify that Tle4 is required for both early acquisition and post-natal stability of corticothalamic neuron-subtype identity. Embryonically, Tle4 promotes acquisition of corticothalamic identity and blocks emergence of core characteristics of subcerebral/corticospinal projection neuron identity, including gene expression and connectivity. During the first post-natal week, when corticothalamic innervation is ongoing, Tle4 is required to stabilize corticothalamic neuron identity, limiting interference from differentiation programs of developmentally related neuron classes. We identify a deacetylation-based epigenetic mechanism by which TLE4 controls Fezf2 expression level by corticothalamic neurons. This contributes to distinction of cortical output subtypes and ensures identity stability for appropriate maturation of corticothalamic neurons.
Collapse
Affiliation(s)
- Maria J Galazo
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - David A Sweetser
- Department of Pediatrics, Divisions of Pediatric Hematology/Oncology and Medical Genetics, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jeffrey D Macklis
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
18
|
Huilgol D, Russ JB, Srivas S, Huang ZJ. The progenitor basis of cortical projection neuron diversity. Curr Opin Neurobiol 2023; 81:102726. [PMID: 37148649 PMCID: PMC10557529 DOI: 10.1016/j.conb.2023.102726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 04/04/2023] [Accepted: 04/09/2023] [Indexed: 05/08/2023]
Abstract
Diverse glutamatergic projection neurons (PNs) mediate myriad processing streams and output channels of the cerebral cortex. Yet, how different types of neural progenitors, such as radial glia (RGs) and intermediate progenitors (IPs), produce PN diversity, and hierarchical organization remains unclear. A fundamental issue is whether RGs constitute a homogeneous, multipotent lineage capable of generating all major PN types through a temporally regulated developmental program, or whether RGs comprise multiple transcriptionally heterogenous pools, each fated to generate a subset of PNs. Beyond RGs, the role of IPs in PN diversification remains underexplored. Addressing these questions requires tracking PN developmental trajectories with cell-type resolution - from transcription factor-defined RGs and IPs to their PN progeny, which are defined not only by laminar location but also by projection patterns and gene expression. Advances in cell-type resolution genetic fate mapping, axon tracing, and spatial transcriptomics may provide the technical capability for answering these fundamental questions.
Collapse
Affiliation(s)
- Dhananjay Huilgol
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Jeffrey B Russ
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Pediatrics, Division of Neurology, Duke University Medical Center, Durham, NC 27710, USA
| | - Sweta Srivas
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Z Josh Huang
- Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Biomedical Engineering, Duke University Pratt School of Engineering, Durham, NC 27708, USA.
| |
Collapse
|
19
|
Felske T, Tocco C, Péron S, Harb K, Alfano C, Galante C, Berninger B, Studer M. Lmo4 synergizes with Fezf2 to promote direct in vivo reprogramming of upper layer cortical neurons and cortical glia towards deep-layer neuron identities. PLoS Biol 2023; 21:e3002237. [PMID: 37552690 PMCID: PMC10409279 DOI: 10.1371/journal.pbio.3002237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 07/06/2023] [Indexed: 08/10/2023] Open
Abstract
In vivo direct neuronal reprogramming relies on the implementation of an exogenous transcriptional program allowing to achieve conversion of a particular neuronal or glial cell type towards a new identity. The transcription factor (TF) Fezf2 is known for its role in neuronal subtype specification of deep-layer (DL) subcortical projection neurons. High ectopic Fezf2 expression in mice can convert both upper-layer (UL) and striatal projection neurons into a corticofugal fate, even if at low efficiency. In this study, we show that Fezf2 synergizes with the nuclear co-adaptor Lmo4 to further enhance reprogramming of UL cortical pyramidal neurons into DL corticofugal neurons, at both embryonic and early postnatal stages. Reprogrammed neurons express DL molecular markers and project toward subcerebral targets, including thalamus, cerebral peduncle (CP), and spinal cord (SC). We also show that co-expression of Fezf2 with the reprogramming factors Neurog2 and Bcl2 in early postnatal mouse glia promotes glia-to-neuron conversion with partial hallmarks of DL neurons and with Lmo4 promoting further morphological complexity. These data support a novel role for Lmo4 in synergizing with Fezf2 during direct lineage conversion in vivo.
Collapse
Affiliation(s)
| | - Chiara Tocco
- Université Côte d’Azur, CNRS, Inserm, iBV, Nice, France
| | - Sophie Péron
- Research Group “Adult Neurogenesis and Cellular Reprogramming”, Institute of Physiological Chemistry, University Medical Center Johannes Gutenberg University, Mainz, Germany
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | - Kawssar Harb
- Université Côte d’Azur, CNRS, Inserm, iBV, Nice, France
| | | | - Chiara Galante
- Research Group “Adult Neurogenesis and Cellular Reprogramming”, Institute of Physiological Chemistry, University Medical Center Johannes Gutenberg University, Mainz, Germany
| | - Benedikt Berninger
- Research Group “Adult Neurogenesis and Cellular Reprogramming”, Institute of Physiological Chemistry, University Medical Center Johannes Gutenberg University, Mainz, Germany
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- Focus Program Translational Neuroscience, Johannes Gutenberg University, Mainz, Germany
| | | |
Collapse
|
20
|
Keijser J, Sprekeler H. Cortical interneurons: fit for function and fit to function? Evidence from development and evolution. Front Neural Circuits 2023; 17:1172464. [PMID: 37215503 PMCID: PMC10192557 DOI: 10.3389/fncir.2023.1172464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 03/30/2023] [Indexed: 05/24/2023] Open
Abstract
Cortical inhibitory interneurons form a broad spectrum of subtypes. This diversity suggests a division of labor, in which each cell type supports a distinct function. In the present era of optimisation-based algorithms, it is tempting to speculate that these functions were the evolutionary or developmental driving force for the spectrum of interneurons we see in the mature mammalian brain. In this study, we evaluated this hypothesis using the two most common interneuron types, parvalbumin (PV) and somatostatin (SST) expressing cells, as examples. PV and SST interneurons control the activity in the cell bodies and the apical dendrites of excitatory pyramidal cells, respectively, due to a combination of anatomical and synaptic properties. But was this compartment-specific inhibition indeed the function for which PV and SST cells originally evolved? Does the compartmental structure of pyramidal cells shape the diversification of PV and SST interneurons over development? To address these questions, we reviewed and reanalyzed publicly available data on the development and evolution of PV and SST interneurons on one hand, and pyramidal cell morphology on the other. These data speak against the idea that the compartment structure of pyramidal cells drove the diversification into PV and SST interneurons. In particular, pyramidal cells mature late, while interneurons are likely committed to a particular fate (PV vs. SST) during early development. Moreover, comparative anatomy and single cell RNA-sequencing data indicate that PV and SST cells, but not the compartment structure of pyramidal cells, existed in the last common ancestor of mammals and reptiles. Specifically, turtle and songbird SST cells also express the Elfn1 and Cbln4 genes that are thought to play a role in compartment-specific inhibition in mammals. PV and SST cells therefore evolved and developed the properties that allow them to provide compartment-specific inhibition before there was selective pressure for this function. This suggest that interneuron diversity originally resulted from a different evolutionary driving force and was only later co-opted for the compartment-specific inhibition it seems to serve in mammals today. Future experiments could further test this idea using our computational reconstruction of ancestral Elfn1 protein sequences.
Collapse
Affiliation(s)
- Joram Keijser
- Modelling of Cognitive Processes, Technical University of Berlin, Berlin, Germany
- Einstein Center for Neurosciences, Charité University Medicine Berlin, Berlin, Germany
| | - Henning Sprekeler
- Modelling of Cognitive Processes, Technical University of Berlin, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Humboldt University of Berlin, Berlin, Germany
| |
Collapse
|
21
|
Song JHT, Ruven C, Patel P, Ding F, Macklis JD, Sahni V. Cbln1 Directs Axon Targeting by Corticospinal Neurons Specifically toward Thoraco-Lumbar Spinal Cord. J Neurosci 2023; 43:1871-1887. [PMID: 36823038 PMCID: PMC10027075 DOI: 10.1523/jneurosci.0710-22.2023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 01/24/2023] [Accepted: 02/02/2023] [Indexed: 02/25/2023] Open
Abstract
Corticospinal neurons (CSN) are centrally required for skilled voluntary movement, which necessitates that they establish precise subcerebral connectivity with the brainstem and spinal cord. However, molecular controls regulating specificity of this projection targeting remain largely unknown. We previously identified that developing CSN subpopulations exhibit striking axon targeting specificity in the spinal white matter. These CSN subpopulations with segmentally distinct spinal projections are also molecularly distinct; a subset of differentially expressed genes between these distinct CSN subpopulations regulate differential axon projection targeting. Rostrolateral CSN extend axons exclusively to bulbar-cervical segments (CSNBC-lat), while caudomedial CSN (CSNmedial) are more heterogeneous, with distinct, intermingled subpopulations extending axons to either bulbar-cervical or thoraco-lumbar segments. Here, we report, in male and female mice, that Cerebellin 1 (Cbln1) is expressed specifically by CSN in medial, but not lateral, sensorimotor cortex. Cbln1 shows highly dynamic temporal expression, with Cbln1 levels in CSN highest during the period of peak axon extension toward thoraco-lumbar segments. Using gain-of-function experiments, we identify that Cbln1 is sufficient to direct thoraco-lumbar axon extension by CSN. Misexpression of Cbln1 in CSNBC-lat either by in utero electroporation, or by postmitotic AAV-mediated gene delivery, redirects these axons past their normal bulbar-cervical targets toward thoracic segments. Further, Cbln1 overexpression in postmitotic CSNBC-lat increases the number of CSNmedial axons that extend past cervical segments into the thoracic cord. Collectively, these results identify that Cbln1 functions as a potent molecular control over thoraco-lumbar CSN axon extension, part of an integrated network of controls over segmentally-specific CSN axon projection targeting.SIGNIFICANCE STATEMENT Corticospinal neurons (CSN) exhibit remarkable diversity and precision of axonal projections to targets in the brainstem and distinct spinal segments; the molecular basis for this targeting diversity is largely unknown. CSN subpopulations projecting to distinct targets are also molecularly distinguishable. Distinct subpopulations degenerate in specific motor neuron diseases, further suggesting that intrinsic molecular differences might underlie differential vulnerability to disease. Here, we identify a novel molecular control, Cbln1, expressed by CSN extending axons to thoraco-lumbar spinal segments. Cbln1 is sufficient, but not required, for CSN axon extension toward distal spinal segments, and Cbln1 expression is controlled by recently identified, CSN-intrinsic regulators of axon extension. Our results identify that Cbln1, together with other regulators, coordinates segmentally precise CSN axon targeting.
Collapse
Affiliation(s)
- Janet H T Song
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, Massachusetts 02138
| | - Carolin Ruven
- Burke Neurological Institute, White Plains, New York 10605
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
| | - Payal Patel
- Burke Neurological Institute, White Plains, New York 10605
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
| | - Frances Ding
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, Massachusetts 02138
| | - Jeffrey D Macklis
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, Massachusetts 02138
| | - Vibhu Sahni
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, Massachusetts 02138
- Burke Neurological Institute, White Plains, New York 10605
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
| |
Collapse
|
22
|
Özel MN, Gibbs CS, Holguera I, Soliman M, Bonneau R, Desplan C. Coordinated control of neuronal differentiation and wiring by sustained transcription factors. Science 2022; 378:eadd1884. [PMID: 36480601 DOI: 10.1126/science.add1884] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The large diversity of cell types in nervous systems presents a challenge in identifying the genetic mechanisms that encode it. Here, we report that nearly 200 distinct neurons in the Drosophila visual system can each be defined by unique combinations of on average 10 continuously expressed transcription factors. We show that targeted modifications of this terminal selector code induce predictable conversions of neuronal fates that appear morphologically and transcriptionally complete. Cis-regulatory analysis of open chromatin links one of these genes to an upstream patterning factor that specifies neuronal fates in stem cells. Experimentally validated network models describe the synergistic regulation of downstream effectors by terminal selectors and ecdysone signaling during brain wiring. Our results provide a generalizable framework of how specific fates are implemented in postmitotic neurons.
Collapse
Affiliation(s)
| | - Claudia Skok Gibbs
- Flatiron Institute, Center for Computational Biology, Simons Foundation, New York, NY 10010, USA.,Center for Data Science, New York University, New York, NY 10003, USA
| | - Isabel Holguera
- Department of Biology, New York University, New York, NY 10003, USA
| | - Mennah Soliman
- Department of Biology, New York University, New York, NY 10003, USA
| | - Richard Bonneau
- Department of Biology, New York University, New York, NY 10003, USA.,Flatiron Institute, Center for Computational Biology, Simons Foundation, New York, NY 10010, USA.,Center for Data Science, New York University, New York, NY 10003, USA
| | - Claude Desplan
- Department of Biology, New York University, New York, NY 10003, USA.,New York University Abu Dhabi, Saadiyat Island, Abu Dhabi, United Arab Emirates
| |
Collapse
|
23
|
Shi X, Zhuang Y, Chen Z, Xu M, Kuang J, Sun XL, Gao L, Kuang X, Zhang H, Li W, Wong SZH, Liu C, Liu L, Jiang D, Pei D, Lin Y, Wu QF. Hierarchical deployment of Tbx3 dictates the identity of hypothalamic KNDy neurons to control puberty onset. SCIENCE ADVANCES 2022; 8:eabq2987. [PMID: 36383654 PMCID: PMC9668310 DOI: 10.1126/sciadv.abq2987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 09/23/2022] [Indexed: 05/17/2023]
Abstract
The neuroendocrine system consists of a heterogeneous collection of neuropeptidergic neurons in the brain, among which hypothalamic KNDy neurons represent an indispensable cell subtype controlling puberty onset. Although neural progenitors and neuronal precursors along the cell lineage hierarchy adopt a cascade diversification strategy to generate hypothalamic neuronal heterogeneity, the cellular logic operating within the lineage to specify a subtype of neuroendocrine neurons remains unclear. As human genetic studies have recently established a link between TBX3 mutations and delayed puberty onset, we systematically studied Tbx3-derived neuronal lineage and Tbx3-dependent neuronal specification and found that Tbx3 hierarchically established and maintained the identity of KNDy neurons for triggering puberty. Apart from the well-established lineage-dependent fate determination, we uncovered rules of interlineage interaction and intralineage retention operating through neuronal differentiation in the absence of Tbx3. Moreover, we revealed that human TBX3 mutations disturbed the phase separation of encoded proteins and impaired transcriptional regulation of key neuropeptides, providing a pathological mechanism underlying TBX3-associated puberty disorders.
Collapse
Affiliation(s)
- Xiang Shi
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Yanrong Zhuang
- IDG/McGovern Institute for Brain Research, Tsinghua–Peking Joint Centre for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Zhenhua Chen
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Mingrui Xu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Junqi Kuang
- University of Chinese Academy of Sciences, Beijing 100101, China
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Xue-Lian Sun
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Lisen Gao
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Xia Kuang
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Huairen Zhang
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Wei Li
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Samuel Zheng Hao Wong
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Chuanyu Liu
- BGI-ShenZhen, Shenzhen 518103, China
- Shenzhen Bay Laboratory, Shenzhen 518000, China
| | - Longqi Liu
- BGI-ShenZhen, Shenzhen 518103, China
- Shenzhen Bay Laboratory, Shenzhen 518000, China
- BGI College and Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Danhua Jiang
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
| | - Duanqing Pei
- Laboratory of Cell Fate Control, School of Life Sciences, Westlake University, Hangzhou 310024, China
| | - Yi Lin
- IDG/McGovern Institute for Brain Research, Tsinghua–Peking Joint Centre for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
- Corresponding author. (Q.-F.W.); (Y.L.)
| | - Qing-Feng Wu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100101, China
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Beijing 100101, China
- Chinese Institute for Brain Research, Beijing 102206, China
- Beijing Children’s Hospital, Capital Medical University, Beijing 100045, China
- Corresponding author. (Q.-F.W.); (Y.L.)
| |
Collapse
|
24
|
Qian Y, Li J, Zhao S, Matthews EA, Adoff M, Zhong W, An X, Yeo M, Park C, Yang X, Wang BS, Southwell DG, Huang ZJ. Programmable RNA sensing for cell monitoring and manipulation. Nature 2022; 610:713-721. [PMID: 36198803 PMCID: PMC10348343 DOI: 10.1038/s41586-022-05280-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 08/26/2022] [Indexed: 12/22/2022]
Abstract
RNA is a central and universal mediator of genetic information underlying the diversity of cell types and cell states, which together shape tissue organization and organismal function across species and lifespans. Despite numerous advances in RNA sequencing technologies and the massive accumulation of transcriptome datasets across the life sciences1,2, the dearth of technologies that use RNAs to observe and manipulate cell types remains a bottleneck in biology and medicine. Here we describe CellREADR (Cell access through RNA sensing by Endogenous ADAR), a programmable RNA-sensing technology that leverages RNA editing mediated by ADAR to couple the detection of cell-defining RNAs with the translation of effector proteins. Viral delivery of CellREADR conferred specific cell-type access in mouse and rat brains and in ex vivo human brain tissues. Furthermore, CellREADR enabled the recording and control of specific types of neurons in behaving mice. CellREADR thus highlights the potential for RNA-based monitoring and editing of animal cells in ways that are specific, versatile, simple and generalizable across organ systems and species, with wide applications in biology, biotechnology and programmable RNA medicine.
Collapse
Affiliation(s)
- Yongjun Qian
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Jiayun Li
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Shengli Zhao
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
| | - Elizabeth A Matthews
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Michael Adoff
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Weixin Zhong
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
| | - Xu An
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
| | - Michele Yeo
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Christine Park
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Xiaolu Yang
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
| | - Bor-Shuen Wang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Derek G Southwell
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
- Department of Neurosurgery, Duke University Medical Center, Durham, NC, USA
| | - Z Josh Huang
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA.
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA.
- Department of Biomedical Engineering, Duke University Pratt School of Engineering, Durham, NC, USA.
| |
Collapse
|
25
|
Carter JA, Strömich L, Peacey M, Chapin SR, Velten L, Steinmetz LM, Brors B, Pinto S, Meyer HV. Transcriptomic diversity in human medullary thymic epithelial cells. Nat Commun 2022; 13:4296. [PMID: 35918316 PMCID: PMC9345899 DOI: 10.1038/s41467-022-31750-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 06/30/2022] [Indexed: 12/03/2022] Open
Abstract
The induction of central T cell tolerance in the thymus depends on the presentation of peripheral self-epitopes by medullary thymic epithelial cells (mTECs). This promiscuous gene expression (pGE) drives mTEC transcriptomic diversity, with non-canonical transcript initiation, alternative splicing, and expression of endogenous retroelements (EREs) representing important but incompletely understood contributors. Here we map the expression of genome-wide transcripts in immature and mature human mTECs using high-throughput 5' cap and RNA sequencing. Both mTEC populations show high splicing entropy, potentially driven by the expression of peripheral splicing factors. During mTEC maturation, rates of global transcript mis-initiation increase and EREs enriched in long terminal repeat retrotransposons are up-regulated, the latter often found in proximity to differentially expressed genes. As a resource, we provide an interactive public interface for exploring mTEC transcriptomic diversity. Our findings therefore help construct a map of transcriptomic diversity in the healthy human thymus and may ultimately facilitate the identification of those epitopes which contribute to autoimmunity and immune recognition of tumor antigens.
Collapse
Affiliation(s)
- Jason A. Carter
- grid.225279.90000 0004 0387 3667Simons Center for Quantitative Biology, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY USA ,grid.36425.360000 0001 2216 9681Medical Scientist Training Program, Stony Brook University, Stony Brook, NY USA ,grid.34477.330000000122986657Department of Surgery, University of Washington, Seattle, WA USA
| | - Léonie Strömich
- grid.7497.d0000 0004 0492 0584German Cancer Research Center, Heidelberg, Germany ,grid.7445.20000 0001 2113 8111Present Address: Imperial College London, London, UK
| | - Matthew Peacey
- grid.225279.90000 0004 0387 3667School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY USA
| | - Sarah R. Chapin
- grid.225279.90000 0004 0387 3667Simons Center for Quantitative Biology, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY USA
| | - Lars Velten
- grid.473715.30000 0004 6475 7299Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain ,grid.5612.00000 0001 2172 2676Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Lars M. Steinmetz
- grid.4709.a0000 0004 0495 846XEuropean Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany ,grid.168010.e0000000419368956Department of Genetics, Stanford University School of Medicine, Stanford, CA USA ,grid.168010.e0000000419368956Stanford Genome Technology Center, Palo Alto, CA USA
| | - Benedikt Brors
- grid.7497.d0000 0004 0492 0584German Cancer Research Center, Heidelberg, Germany
| | - Sheena Pinto
- grid.7497.d0000 0004 0492 0584German Cancer Research Center, Heidelberg, Germany
| | - Hannah V. Meyer
- grid.225279.90000 0004 0387 3667Simons Center for Quantitative Biology, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY USA
| |
Collapse
|
26
|
Nano PR, Bhaduri A. Evaluation of advances in cortical development using model systems. Dev Neurobiol 2022; 82:408-427. [PMID: 35644985 PMCID: PMC10924780 DOI: 10.1002/dneu.22879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 04/26/2022] [Accepted: 04/30/2022] [Indexed: 11/11/2022]
Abstract
Compared with that of even the closest primates, the human cortex displays a high degree of specialization and expansion that largely emerges developmentally. Although decades of research in the mouse and other model systems has revealed core tenets of cortical development that are well preserved across mammalian species, small deviations in transcription factor expression, novel cell types in primates and/or humans, and unique cortical architecture distinguish the human cortex. Importantly, many of the genes and signaling pathways thought to drive human-specific cortical expansion also leave the brain vulnerable to disease, as the misregulation of these factors is highly correlated with neurodevelopmental and neuropsychiatric disorders. However, creating a comprehensive understanding of human-specific cognition and disease remains challenging. Here, we review key stages of cortical development and highlight known or possible differences between model systems and the developing human brain. By identifying the developmental trajectories that may facilitate uniquely human traits, we highlight open questions in need of approaches to examine these processes in a human context and reveal translatable insights into human developmental disorders.
Collapse
Affiliation(s)
- Patricia R Nano
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Aparna Bhaduri
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
27
|
Romero-Morales AI, Gama V. Revealing the Impact of Mitochondrial Fitness During Early Neural Development Using Human Brain Organoids. Front Mol Neurosci 2022; 15:840265. [PMID: 35571368 PMCID: PMC9102998 DOI: 10.3389/fnmol.2022.840265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Mitochondrial homeostasis -including function, morphology, and inter-organelle communication- provides guidance to the intrinsic developmental programs of corticogenesis, while also being responsive to environmental and intercellular signals. Two- and three-dimensional platforms have become useful tools to interrogate the capacity of cells to generate neuronal and glia progeny in a background of metabolic dysregulation, but the mechanistic underpinnings underlying the role of mitochondria during human neurogenesis remain unexplored. Here we provide a concise overview of cortical development and the use of pluripotent stem cell models that have contributed to our understanding of mitochondrial and metabolic regulation of early human brain development. We finally discuss the effects of mitochondrial fitness dysregulation seen under stress conditions such as metabolic dysregulation, absence of developmental apoptosis, and hypoxia; and the avenues of research that can be explored with the use of brain organoids.
Collapse
Affiliation(s)
| | - Vivian Gama
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
28
|
Herrera E, Escalante A. Transcriptional Control of Axon Guidance at Midline Structures. Front Cell Dev Biol 2022; 10:840005. [PMID: 35265625 PMCID: PMC8900194 DOI: 10.3389/fcell.2022.840005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
The development of the nervous system is a time-ordered and multi-stepped process that includes neurogenesis and neuronal specification, axonal navigation, and circuits assembly. During axonal navigation, the growth cone, a dynamic structure located at the tip of the axon, senses environmental signals that guide axons towards their final targets. The expression of a specific repertoire of receptors on the cell surface of the growth cone together with the activation of a set of intracellular transducing molecules, outlines the response of each axon to specific guidance cues. This collection of axon guidance molecules is defined by the transcriptome of the cell which, in turn, depends on transcriptional and epigenetic regulators that modify the structure and DNA accessibility to determine what genes will be expressed to elicit specific axonal behaviors. Studies focused on understanding how axons navigate intermediate targets, such as the floor plate of vertebrates or the mammalian optic chiasm, have largely contributed to our knowledge of how neurons wire together during development. In fact, investigations on axon navigation at these midline structures led to the identification of many of the currently known families of proteins that act as guidance cues and their corresponding receptors. Although the transcription factors and the regulatory mechanisms that control the expression of these molecules are not well understood, important advances have been made in recent years in this regard. Here we provide an updated overview on the current knowledge about the transcriptional control of axon guidance and the selection of trajectories at midline structures.
Collapse
|
29
|
Deng H, Xiao X, Yang T, Ritola K, Hantman A, Li Y, Huang ZJ, Li B. A genetically defined insula-brainstem circuit selectively controls motivational vigor. Cell 2021; 184:6344-6360.e18. [PMID: 34890577 PMCID: PMC9103523 DOI: 10.1016/j.cell.2021.11.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/18/2021] [Accepted: 11/10/2021] [Indexed: 12/24/2022]
Abstract
The anterior insular cortex (aIC) plays a critical role in cognitive and motivational control of behavior, but the underlying neural mechanism remains elusive. Here, we show that aIC neurons expressing Fezf2 (aICFezf2), which are the pyramidal tract neurons, signal motivational vigor and invigorate need-seeking behavior through projections to the brainstem nucleus tractus solitarii (NTS). aICFezf2 neurons and their postsynaptic NTS neurons acquire anticipatory activity through learning, which encodes the perceived value and the vigor of actions to pursue homeostatic needs. Correspondingly, aIC → NTS circuit activity controls vigor, effort, and striatal dopamine release but only if the action is learned and the outcome is needed. Notably, aICFezf2 neurons do not represent taste or valence. Moreover, aIC → NTS activity neither drives reinforcement nor influences total consumption. These results pinpoint specific functions of aIC → NTS circuit for selectively controlling motivational vigor and suggest that motivation is subserved, in part, by aIC's top-down regulation of dopamine signaling.
Collapse
Affiliation(s)
- Hanfei Deng
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| | - Xiong Xiao
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Tao Yang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | | | - Adam Hantman
- HHMI Janelia Research Campus, Ashburn, VA 20147, USA
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Beijing 100871, China
| | - Z Josh Huang
- Department of Neurobiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Bo Li
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
30
|
Mirabella F, Desiato G, Mancinelli S, Fossati G, Rasile M, Morini R, Markicevic M, Grimm C, Amegandjin C, Termanini A, Peano C, Kunderfranco P, di Cristo G, Zerbi V, Menna E, Lodato S, Matteoli M, Pozzi D. Prenatal interleukin 6 elevation increases glutamatergic synapse density and disrupts hippocampal connectivity in offspring. Immunity 2021; 54:2611-2631.e8. [PMID: 34758338 PMCID: PMC8585508 DOI: 10.1016/j.immuni.2021.10.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/24/2021] [Accepted: 10/07/2021] [Indexed: 02/07/2023]
Abstract
Early prenatal inflammatory conditions are thought to be a risk factor for different neurodevelopmental disorders. Maternal interleukin-6 (IL-6) elevation during pregnancy causes abnormal behavior in offspring, but whether these defects result from altered synaptic developmental trajectories remains unclear. Here we showed that transient IL-6 elevation via injection into pregnant mice or developing embryos enhanced glutamatergic synapses and led to overall brain hyperconnectivity in offspring into adulthood. IL-6 activated synaptogenesis gene programs in glutamatergic neurons and required the transcription factor STAT3 and expression of the RGS4 gene. The STAT3-RGS4 pathway was also activated in neonatal brains during poly(I:C)-induced maternal immune activation, which mimics viral infection during pregnancy. These findings indicate that IL-6 elevation at early developmental stages is sufficient to exert a long-lasting effect on glutamatergic synaptogenesis and brain connectivity, providing a mechanistic framework for the association between prenatal inflammatory events and brain neurodevelopmental disorders.
Collapse
Affiliation(s)
- Filippo Mirabella
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Genni Desiato
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Institute of Neuroscience - National Research Council, 20139 Milan, Italy
| | - Sara Mancinelli
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Giuliana Fossati
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Marco Rasile
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy
| | - Raffaella Morini
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Marija Markicevic
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich 8057, Switzerland
| | - Christina Grimm
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich 8057, Switzerland
| | - Clara Amegandjin
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada; CHU Sainte-Justine Research Center, Montréal, QC, Canada
| | - Alberto Termanini
- Bioinformatic Unit, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Clelia Peano
- Institute of Genetic and Biomedical Research, UoS Milan, National Research Council, 20089 Rozzano, Milan, Italy; Genomic Unit, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Paolo Kunderfranco
- Bioinformatic Unit, Humanitas Clinical and Research Center, 20089 Rozzano, Milan, Italy
| | - Graziella di Cristo
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada; CHU Sainte-Justine Research Center, Montréal, QC, Canada
| | - Valerio Zerbi
- Neuroscience Center Zürich, ETH Zürich and University of Zürich, Zürich 8057, Switzerland; Neural Control of Movement Lab, Department of Health Sciences and Technology, ETH Zürich, Zürich 8057, Switzerland
| | - Elisabetta Menna
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Institute of Neuroscience - National Research Council, 20139 Milan, Italy
| | - Simona Lodato
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Michela Matteoli
- IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy; Institute of Neuroscience - National Research Council, 20139 Milan, Italy.
| | - Davide Pozzi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20090 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Rozzano, Milan, Italy.
| |
Collapse
|
31
|
A new player in neural circuits of emotions. Nat Neurosci 2021; 24:1506-1507. [PMID: 34654921 DOI: 10.1038/s41593-021-00945-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
32
|
Genetically identified amygdala-striatal circuits for valence-specific behaviors. Nat Neurosci 2021; 24:1586-1600. [PMID: 34663958 PMCID: PMC8556347 DOI: 10.1038/s41593-021-00927-0] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/25/2021] [Indexed: 11/08/2022]
Abstract
The basolateral amygdala (BLA) plays essential roles in behaviors motivated by stimuli with either positive or negative valence, but how it processes motivationally opposing information and participates in establishing valence-specific behaviors remains unclear. Here, by targeting Fezf2-expressing neurons in the BLA, we identify and characterize two functionally distinct classes in behaving mice, the negative-valence neurons and positive-valence neurons, which innately represent aversive and rewarding stimuli, respectively, and through learning acquire predictive responses that are essential for punishment avoidance or reward seeking. Notably, these two classes of neurons receive inputs from separate sets of sensory and limbic areas, and convey punishment and reward information through projections to the nucleus accumbens and olfactory tubercle, respectively, to drive negative and positive reinforcement. Thus, valence-specific BLA neurons are wired with distinctive input-output structures, forming a circuit framework that supports the roles of the BLA in encoding, learning and executing valence-specific motivated behaviors.
Collapse
|
33
|
Sahni V, Shnider SJ, Jabaudon D, Song JHT, Itoh Y, Greig LC, Macklis JD. Corticospinal neuron subpopulation-specific developmental genes prospectively indicate mature segmentally specific axon projection targeting. Cell Rep 2021; 37:109843. [PMID: 34686320 PMCID: PMC8653526 DOI: 10.1016/j.celrep.2021.109843] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 05/27/2021] [Accepted: 09/26/2021] [Indexed: 11/11/2022] Open
Abstract
For precise motor control, distinct subpopulations of corticospinal neurons (CSN) must extend axons to distinct spinal segments, from proximal targets in the brainstem and cervical cord to distal targets in thoracic and lumbar spinal segments. We find that developing CSN subpopulations exhibit striking axon targeting specificity in spinal white matter, which establishes the foundation for durable specificity of adult corticospinal circuitry. Employing developmental retrograde and anterograde labeling, and their distinct neocortical locations, we purified developing CSN subpopulations using fluorescence-activated cell sorting to identify genes differentially expressed between bulbar-cervical and thoracolumbar-projecting CSN subpopulations at critical developmental times. These segmentally distinct CSN subpopulations are molecularly distinct from the earliest stages of axon extension, enabling prospective identification even before eventual axon targeting decisions are evident in the spinal cord. This molecular delineation extends beyond simple spatial separation of these subpopulations in the cortex. Together, these results identify candidate molecular controls over segmentally specific corticospinal axon projection targeting.
Collapse
Affiliation(s)
- Vibhu Sahni
- Department of Stem Cell and Regenerative Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Sara J Shnider
- Department of Stem Cell and Regenerative Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Denis Jabaudon
- Department of Stem Cell and Regenerative Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Janet H T Song
- Department of Stem Cell and Regenerative Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Yasuhiro Itoh
- Department of Stem Cell and Regenerative Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Luciano C Greig
- Department of Stem Cell and Regenerative Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Jeffrey D Macklis
- Department of Stem Cell and Regenerative Biology, Center for Brain Science, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
34
|
Sahni V, Itoh Y, Shnider SJ, Macklis JD. Crim1 and Kelch-like 14 exert complementary dual-directional developmental control over segmentally specific corticospinal axon projection targeting. Cell Rep 2021; 37:109842. [PMID: 34686337 PMCID: PMC8697027 DOI: 10.1016/j.celrep.2021.109842] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 05/27/2021] [Accepted: 09/26/2021] [Indexed: 11/20/2022] Open
Abstract
The cerebral cortex executes highly skilled movement, necessitating that it connects accurately with specific brainstem and spinal motor circuitry. Corticospinal neurons (CSN) must correctly target specific spinal segments, but the basis for this targeting remains unknown. In the accompanying report, we show that segmentally distinct CSN subpopulations are molecularly distinct from early development, identifying candidate molecular controls over segmentally specific axon targeting. Here, we functionally investigate two of these candidate molecular controls, Crim1 and Kelch-like 14 (Klhl14), identifying their critical roles in directing CSN axons to appropriate spinal segmental levels in the white matter prior to axon collateralization. Crim1 and Klhl14 are specifically expressed by distinct CSN subpopulations and regulate their differental white matter projection targeting-Crim1 directs thoracolumbar axon extension, while Klhl14 limits axon extension to bulbar-cervical segments. These molecular regulators of descending spinal projections constitute the first stages of a dual-directional set of complementary controls over CSN diversity for segmentally and functionally distinct circuitry.
Collapse
Affiliation(s)
- Vibhu Sahni
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Yasuhiro Itoh
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Sara J Shnider
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Jeffrey D Macklis
- Department of Stem Cell and Regenerative Biology, and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
35
|
Matho KS, Huilgol D, Galbavy W, He M, Kim G, An X, Lu J, Wu P, Di Bella DJ, Shetty AS, Palaniswamy R, Hatfield J, Raudales R, Narasimhan A, Gamache E, Levine JM, Tucciarone J, Szelenyi E, Harris JA, Mitra PP, Osten P, Arlotta P, Huang ZJ. Genetic dissection of the glutamatergic neuron system in cerebral cortex. Nature 2021; 598:182-187. [PMID: 34616069 PMCID: PMC8494647 DOI: 10.1038/s41586-021-03955-9] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 08/25/2021] [Indexed: 11/09/2022]
Abstract
Diverse types of glutamatergic pyramidal neurons mediate the myriad processing streams and output channels of the cerebral cortex1,2, yet all derive from neural progenitors of the embryonic dorsal telencephalon3,4. Here we establish genetic strategies and tools for dissecting and fate-mapping subpopulations of pyramidal neurons on the basis of their developmental and molecular programs. We leverage key transcription factors and effector genes to systematically target temporal patterning programs in progenitors and differentiation programs in postmitotic neurons. We generated over a dozen temporally inducible mouse Cre and Flp knock-in driver lines to enable the combinatorial targeting of major progenitor types and projection classes. Combinatorial strategies confer viral access to subsets of pyramidal neurons defined by developmental origin, marker expression, anatomical location and projection targets. These strategies establish an experimental framework for understanding the hierarchical organization and developmental trajectory of subpopulations of pyramidal neurons that assemble cortical processing networks and output channels.
Collapse
Affiliation(s)
- Katherine S Matho
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Dhananjay Huilgol
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
| | - William Galbavy
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Program in Neuroscience, Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, USA
| | - Miao He
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Gukhan Kim
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Xu An
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
| | - Jiangteng Lu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Shanghai Jiaotong University Medical School, Shanghai, China
| | - Priscilla Wu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Daniela J Di Bella
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Ashwin S Shetty
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | | | - Joshua Hatfield
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
| | - Ricardo Raudales
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Program in Neuroscience, Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, USA
| | - Arun Narasimhan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Eric Gamache
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Jesse M Levine
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Program in Neuroscience and Medical Scientist Training Program, Stony Brook University, New York, NY, USA
| | - Jason Tucciarone
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Program in Neuroscience and Medical Scientist Training Program, Stony Brook University, New York, NY, USA
- Department of Psychiatry, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Eric Szelenyi
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Julie A Harris
- Program in Neuroscience and Medical Scientist Training Program, Stony Brook University, New York, NY, USA
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Partha P Mitra
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Pavel Osten
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Paola Arlotta
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Z Josh Huang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA.
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
36
|
Lattke M, Goldstone R, Ellis JK, Boeing S, Jurado-Arjona J, Marichal N, MacRae JI, Berninger B, Guillemot F. Extensive transcriptional and chromatin changes underlie astrocyte maturation in vivo and in culture. Nat Commun 2021; 12:4335. [PMID: 34267208 PMCID: PMC8282848 DOI: 10.1038/s41467-021-24624-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 06/16/2021] [Indexed: 11/08/2022] Open
Abstract
Astrocytes have essential functions in brain homeostasis that are established late in differentiation, but the mechanisms underlying the functional maturation of astrocytes are not well understood. Here we identify extensive transcriptional changes that occur during murine astrocyte maturation in vivo that are accompanied by chromatin remodelling at enhancer elements. Investigating astrocyte maturation in a cell culture model revealed that in vitro-differentiated astrocytes lack expression of many mature astrocyte-specific genes, including genes for the transcription factors Rorb, Dbx2, Lhx2 and Fezf2. Forced expression of these factors in vitro induces distinct sets of mature astrocyte-specific transcripts. Culturing astrocytes in a three-dimensional matrix containing FGF2 induces expression of Rorb, Dbx2 and Lhx2 and improves astrocyte maturity based on transcriptional and chromatin profiles. Therefore, extrinsic signals orchestrate the expression of multiple intrinsic regulators, which in turn induce in a modular manner the transcriptional and chromatin changes underlying astrocyte maturation.
Collapse
Affiliation(s)
- Michael Lattke
- Neural Stem Cell Biology Laboratory, The Francis Crick Institute, London, UK
| | - Robert Goldstone
- Advanced Sequencing Facility, The Francis Crick Institute, London, UK
| | - James K Ellis
- Metabolomics Science Technology Platform, The Francis Crick Institute, London, UK
| | - Stefan Boeing
- Software Development & Machine Learning Team, The Francis Crick Institute, London, UK
- Bioinformatics & Biostatistics, The Francis Crick Institute, London, UK
| | - Jerónimo Jurado-Arjona
- Institute of Psychiatry, Psychology & Neuroscience, Centre for Developmental Neurobiology, King's College London, London, UK
| | - Nicolás Marichal
- Institute of Psychiatry, Psychology & Neuroscience, Centre for Developmental Neurobiology, King's College London, London, UK
| | - James I MacRae
- Metabolomics Science Technology Platform, The Francis Crick Institute, London, UK
| | - Benedikt Berninger
- Institute of Psychiatry, Psychology & Neuroscience, Centre for Developmental Neurobiology, King's College London, London, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- The Francis Crick Institute, London, UK
| | - Francois Guillemot
- Neural Stem Cell Biology Laboratory, The Francis Crick Institute, London, UK.
| |
Collapse
|
37
|
Di Bella DJ, Habibi E, Stickels RR, Scalia G, Brown J, Yadollahpour P, Yang SM, Abbate C, Biancalani T, Macosko EZ, Chen F, Regev A, Arlotta P. Molecular logic of cellular diversification in the mouse cerebral cortex. Nature 2021; 595:554-559. [PMID: 34163074 PMCID: PMC9006333 DOI: 10.1038/s41586-021-03670-5] [Citation(s) in RCA: 235] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 05/24/2021] [Indexed: 02/06/2023]
Abstract
The mammalian cerebral cortex has an unparalleled diversity of cell types, which are generated during development through a series of temporally orchestrated events that are under tight evolutionary constraint and are critical for proper cortical assembly and function1,2. However, the molecular logic that governs the establishment and organization of cortical cell types remains unknown, largely due to the large number of cell classes that undergo dynamic cell-state transitions over extended developmental timelines. Here we generate a comprehensive atlas of the developing mouse neocortex, using single-cell RNA sequencing and single-cell assay for transposase-accessible chromatin using sequencing. We sampled the neocortex every day throughout embryonic corticogenesis and at early postnatal ages, and complemented the sequencing data with a spatial transcriptomics time course. We computationally reconstruct developmental trajectories across the diversity of cortical cell classes, and infer their spatial organization and the gene regulatory programs that accompany their lineage bifurcation decisions and differentiation trajectories. Finally, we demonstrate how this developmental map pinpoints the origin of lineage-specific developmental abnormalities that are linked to aberrant corticogenesis in mutant mice. The data provide a global picture of the regulatory mechanisms that govern cellular diversification in the neocortex.
Collapse
Affiliation(s)
- Daniela J. Di Bella
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ehsan Habibi
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA,Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Gabriele Scalia
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA,Current address: Genentech, South San Francisco, CA, USA
| | - Juliana Brown
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Payman Yadollahpour
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sung Min Yang
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Catherine Abbate
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Tommasso Biancalani
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA,Current address: Genentech, South San Francisco, CA, USA
| | - Evan Z. Macosko
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Fei Chen
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA,Current address: Genentech, South San Francisco, CA, USA,Howard Hughes Medical Institute, Koch Institute of Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA,Correspondence should be addressed to and
| | - Paola Arlotta
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA,Correspondence should be addressed to and
| |
Collapse
|
38
|
Tsyporin J, Tastad D, Ma X, Nehme A, Finn T, Huebner L, Liu G, Gallardo D, Makhamreh A, Roberts JM, Katzman S, Sestan N, McConnell SK, Yang Z, Qiu S, Chen B. Transcriptional repression by FEZF2 restricts alternative identities of cortical projection neurons. Cell Rep 2021; 35:109269. [PMID: 34161768 PMCID: PMC8327856 DOI: 10.1016/j.celrep.2021.109269] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/05/2021] [Accepted: 05/27/2021] [Indexed: 11/20/2022] Open
Abstract
Projection neuron subtype identities in the cerebral cortex are established by expressing pan-cortical and subtype-specific effector genes that execute terminal differentiation programs bestowing neurons with a glutamatergic neuron phenotype and subtype-specific morphology, physiology, and axonal projections. Whether pan-cortical glutamatergic and subtype-specific characteristics are regulated by the same genes or controlled by distinct programs remains largely unknown. Here, we show that FEZF2 functions as a transcriptional repressor, and it regulates subtype-specific identities of both corticothalamic and subcerebral neurons by selectively repressing expression of genes inappropriate for each neuronal subtype. We report that TLE4, specifically expressed in layer 6 corticothalamic neurons, is recruited by FEZF2 to inhibit layer 5 subcerebral neuronal genes. Together with previous studies, our results indicate that a cortical glutamatergic identity is specified by multiple parallel pathways active in progenitor cells, whereas projection neuron subtype-specific identity is achieved through selectively repressing genes associated with alternate identities in differentiating neurons.
Collapse
Affiliation(s)
- Jeremiah Tsyporin
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
| | - David Tastad
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
| | - Xiaokuang Ma
- Department of Basic Medical Sciences, University of Arizona College of Medicine - Phoenix, Phoenix, AZ 85004, USA
| | - Antoine Nehme
- Department of Basic Medical Sciences, University of Arizona College of Medicine - Phoenix, Phoenix, AZ 85004, USA
| | - Thomas Finn
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
| | - Liora Huebner
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
| | - Guoping Liu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute for Translational Brain Research, Institutes of Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Daisy Gallardo
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
| | - Amr Makhamreh
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
| | - Jacqueline M Roberts
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
| | - Solomon Katzman
- Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Nenad Sestan
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06520, USA
| | | | - Zhengang Yang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute for Translational Brain Research, Institutes of Brain Science, Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Shenfeng Qiu
- Department of Basic Medical Sciences, University of Arizona College of Medicine - Phoenix, Phoenix, AZ 85004, USA
| | - Bin Chen
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA.
| |
Collapse
|
39
|
Fenlon LR, Suarez R, Lynton Z, Richards LJ. The evolution, formation and connectivity of the anterior commissure. Semin Cell Dev Biol 2021; 118:50-59. [PMID: 33958283 DOI: 10.1016/j.semcdb.2021.04.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/08/2021] [Accepted: 04/10/2021] [Indexed: 10/21/2022]
Abstract
The anterior commissure is the most ancient of the forebrain interhemispheric connections among all vertebrates. Indeed, it is the predominant pallial commissure in all non-eutherian vertebrates, universally subserving basic functions related to olfaction and survival. A key feature of the anterior commissure is its ability to convey connections from diverse brain areas, such as most of the neocortex in non-eutherian mammals, thereby mediating the bilateral integration of diverse functions. Shared developmental mechanisms between the anterior commissure and more evolutionarily recent commissures, such as the corpus callosum in eutherians, have led to the hypothesis that the former may have been a precursor for additional expansion of commissural circuits. However, differences between the formation of the anterior commissure and other telencephalic commissures suggest that independent developmental mechanisms underlie the emergence of these connections in extant species. Here, we review the developmental mechanisms and connectivity of the anterior commissure across evolutionarily distant species, and highlight its potential functional importance in humans, both in the course of normal neurodevelopment, and as a site of plastic axonal rerouting in the absence or damage of other connections.
Collapse
Affiliation(s)
- Laura R Fenlon
- The University of Queensland, The Queensland Brain Institute, Brisbane, Australia.
| | - Rodrigo Suarez
- The University of Queensland, The Queensland Brain Institute, Brisbane, Australia
| | - Zorana Lynton
- The University of Queensland, The Queensland Brain Institute, Brisbane, Australia; The Faculty of Medicine, Brisbane, Australia
| | - Linda J Richards
- The University of Queensland, The Queensland Brain Institute, Brisbane, Australia; The School of Biomedical Sciences, Brisbane, Australia.
| |
Collapse
|
40
|
Herrero-Navarro Á, Puche-Aroca L, Moreno-Juan V, Sempere-Ferràndez A, Espinosa A, Susín R, Torres-Masjoan L, Leyva-Díaz E, Karow M, Figueres-Oñate M, López-Mascaraque L, López-Atalaya JP, Berninger B, López-Bendito G. Astrocytes and neurons share region-specific transcriptional signatures that confer regional identity to neuronal reprogramming. SCIENCE ADVANCES 2021; 7:7/15/eabe8978. [PMID: 33827819 PMCID: PMC8026135 DOI: 10.1126/sciadv.abe8978] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 02/18/2021] [Indexed: 05/15/2023]
Abstract
Neural cell diversity is essential to endow distinct brain regions with specific functions. During development, progenitors within these regions are characterized by specific gene expression programs, contributing to the generation of diversity in postmitotic neurons and astrocytes. While the region-specific molecular diversity of neurons and astrocytes is increasingly understood, whether these cells share region-specific programs remains unknown. Here, we show that in the neocortex and thalamus, neurons and astrocytes express shared region-specific transcriptional and epigenetic signatures. These signatures not only distinguish cells across these two brain regions but are also detected across substructures within regions, such as distinct thalamic nuclei, where clonal analysis reveals the existence of common nucleus-specific progenitors for neurons and astrocytes. Consistent with their shared molecular signature, regional specificity is maintained following astrocyte-to-neuron reprogramming. A detailed understanding of these regional-specific signatures may thus inform strategies for future cell-based brain repair.
Collapse
Affiliation(s)
- Álvaro Herrero-Navarro
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Lorenzo Puche-Aroca
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Verónica Moreno-Juan
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Alejandro Sempere-Ferràndez
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Ana Espinosa
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Rafael Susín
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Laia Torres-Masjoan
- Institute of Psychiatry, Psychology, and Neuroscience, Centre for Developmental Neurobiology, and MRC Centre for Neurodevelopmental Disorders, King's College London, SE1 1UL London, UK
| | - Eduardo Leyva-Díaz
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Marisa Karow
- Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany
- Physiological Genomics, Biomedical Center, Ludwig Maximilians University Munich, 82152 Planegg/Munich, Germany
| | | | | | - José P López-Atalaya
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain
| | - Benedikt Berninger
- Institute of Psychiatry, Psychology, and Neuroscience, Centre for Developmental Neurobiology, and MRC Centre for Neurodevelopmental Disorders, King's College London, SE1 1UL London, UK
- Institute of Physiological Chemistry, University Medical Center Johannes Gutenberg University Mainz, 55128 Mainz, Germany
| | - Guillermina López-Bendito
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d'Alacant, Spain.
| |
Collapse
|
41
|
Li J, Sun L, Peng XL, Yu XM, Qi SJ, Lu ZJ, Han JDJ, Shen Q. Integrative genomic analysis of early neurogenesis reveals a temporal genetic program for differentiation and specification of preplate and Cajal-Retzius neurons. PLoS Genet 2021; 17:e1009355. [PMID: 33760820 PMCID: PMC7990179 DOI: 10.1371/journal.pgen.1009355] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 01/12/2021] [Indexed: 01/02/2023] Open
Abstract
Neurogenesis in the developing neocortex begins with the generation of the preplate, which consists of early-born neurons including Cajal-Retzius (CR) cells and subplate neurons. Here, utilizing the Ebf2-EGFP transgenic mouse in which EGFP initially labels the preplate neurons then persists in CR cells, we reveal the dynamic transcriptome profiles of early neurogenesis and CR cell differentiation. Genome-wide RNA-seq and ChIP-seq analyses at multiple early neurogenic stages have revealed the temporal gene expression dynamics of early neurogenesis and distinct histone modification patterns in early differentiating neurons. We have identified a new set of coding genes and lncRNAs involved in early neuronal differentiation and validated with functional assays in vitro and in vivo. In addition, at E15.5 when Ebf2-EGFP+ cells are mostly CR neurons, single-cell sequencing analysis of purified Ebf2-EGFP+ cells uncovers molecular heterogeneities in CR neurons, but without apparent clustering of cells with distinct regional origins. Along a pseudotemporal trajectory these cells are classified into three different developing states, revealing genetic cascades from early generic neuronal differentiation to late fate specification during the establishment of CR neuron identity and function. Our findings shed light on the molecular mechanisms governing the early differentiation steps during cortical development, especially CR neuron differentiation. Neural stem cells and progenitor cells in the embryonic brain give rise to neurons following a precise temporal order after initial expansion. Early-born neurons including Cajal-Retzius (CR) cells and subplate neurons form the preplate in the developing cerebral cortex, then CR neurons occupy the layer 1, playing an important role in cortical histogenesis. The molecular mechanisms governing the early neuronal differentiation processes remain to be explored. Here, by genome-wide approaches including bulk RNA-seq, single-cell RNA-seq and ChIP-seq, we comprehensively characterized the temporal dynamic gene expression profile and epigenetic status at different stages during early cortical development and uncovered molecularly heterogeneous subpopulations within the CR cells. We revealed CR neuron signatures and cell type-specific histone modification patterns along early neuron specification. Using in vitro and in vivo assays, we identified novel lncRNAs as potential functional regulators in preplate differentiation and CR neuron identity establishment. Our study provides a comprehensive analysis of the genetic and epigenetic programs during neuronal differentiation and would help bring new insights into the early cortical neurogenesis process, particularly the differentiation of CR neurons.
Collapse
Affiliation(s)
- Jia Li
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
- PTN graduate program, School of Life Sciences, Peking University, Beijing, China
- School of Medicine, Tsinghua University, Beijing, China
| | - Lei Sun
- PTN graduate program, School of Life Sciences, Tsinghua University, Beijing, China
| | | | - Xiao-Ming Yu
- School of Medicine, Tsinghua University, Beijing, China
| | - Shao-Jun Qi
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
- School of Medicine, Tsinghua University, Beijing, China
| | - Zhi John Lu
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Jing-Dong J. Han
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qin Shen
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Frontier Science Center for Stem Cell Research, Ministry of Education, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Brain and Spinal Cord Clinical Research Center, Tongji University, Shanghai, China
- * E-mail:
| |
Collapse
|
42
|
Hou Y, Zhang Q, Liu H, Wu J, Shi Y, Qi Y, Shao M, Yang Z, Lu J, Wu Z, Gong L, He M. Topographical organization of mammillary neurogenesis and efferent projections in the mouse brain. Cell Rep 2021; 34:108712. [PMID: 33567285 DOI: 10.1016/j.celrep.2021.108712] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 12/01/2020] [Accepted: 01/12/2021] [Indexed: 01/23/2023] Open
Abstract
The mammillary body is a hypothalamic nucleus that has important functions in memory and spatial navigation, but its developmental principles remain not well understood. Here, we identify progenitor-specific Fezf2 expression in the developing mammillary body and develop an intersectional fate-mapping approach to demonstrate that Fezf2+ mammillary progenitors generate mammillary neurons in a rostral-dorsal-lateral to caudal-ventral-medial fashion. Axonal tracing from different temporal cohorts of labeled mammillary neurons reveal their topographical organization. Unsupervised hierarchical clustering based on intrinsic properties further identify two distinct neuronal clusters independent of birthdates in the medial nuclei. In addition, we generate Fezf2 knockout mice and observe the smaller mammillary body with largely normal anatomy and mildly affected cellular electrophysiology, in contrast to more severe deficits in neuronal differentiation and projection in many other brain regions. These results indicate that Fezf2 may function differently in the mammillary body. Our results provide important insights for mammillary development and connectivity.
Collapse
Affiliation(s)
- Yongjie Hou
- Institutes of Brain Science, Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qi Zhang
- Institutes of Brain Science, Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hongzhi Liu
- Institutes of Brain Science, Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jinyun Wu
- Institutes of Brain Science, Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun Shi
- Institutes of Brain Science, Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yanqing Qi
- Institutes of Brain Science, Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mengmeng Shao
- Department of Anatomy and Physiology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhengang Yang
- Institutes of Brain Science, Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiangteng Lu
- Department of Anatomy and Physiology, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhuhao Wu
- Department of Cell, Developmental and Regenerative Biology, Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ling Gong
- Institutes of Brain Science, Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Miao He
- Institutes of Brain Science, Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
43
|
Ji Y, Zhang X, Wang Z, Qin W, Liu H, Xue K, Tang J, Xu Q, Zhu D, Liu F, Yu C. Genes associated with gray matter volume alterations in schizophrenia. Neuroimage 2020; 225:117526. [PMID: 33147509 DOI: 10.1016/j.neuroimage.2020.117526] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 10/28/2020] [Indexed: 12/11/2022] Open
Abstract
Although both schizophrenia and gray matter volume (GMV) show high heritability, however, genes accounting for GMV alterations in schizophrenia remain largely unknown. Based on risk genes identified in schizophrenia by the genome-wide association study of the Schizophrenia Working Group of the Psychiatric Genomics Consortium, we used transcription-neuroimaging association analysis to test that which of these genes are associated with GMV changes in schizophrenia. For each brain tissue sample, the expression profiles of 196 schizophrenia risk genes were extracted from six donated normal brains of the Allen Human Brain Atlas, and GMV differences between patients with schizophrenia and healthy controls were calculated based on five independent case-control structural MRI datasets (276 patients and 284 controls). Genes associated with GMV changes in schizophrenia were identified by performing cross-sample spatial correlations between expression levels of each gene and case-control GMV difference derived from the five MRI datasets integrated by harmonization and meta-analysis. We found that expression levels of 98 genes consistently showed significant cross-sample spatial correlations with GMV changes in schizophrenia. These genes were functionally enriched for chemical synaptic transmission, central nervous system development, and cell projection. Overall, this study provides a set of genes possibly associated with GMV changes in schizophrenia, which could be used as candidate genes to explore biological mechanisms underlying the structural impairments in schizophrenia.
Collapse
Affiliation(s)
- Yuan Ji
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xue Zhang
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zirui Wang
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Wen Qin
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Huaigui Liu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Kaizhong Xue
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jie Tang
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Qiang Xu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Dan Zhu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Feng Liu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Chunshui Yu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
44
|
An evolutionarily acquired microRNA shapes development of mammalian cortical projections. Proc Natl Acad Sci U S A 2020; 117:29113-29122. [PMID: 33139574 PMCID: PMC7682328 DOI: 10.1073/pnas.2006700117] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The mammalian central nervous system contains unique projections from the cerebral cortex thought to underpin complex motor and cognitive skills, including the corticospinal tract and corpus callosum. The neurons giving rise to these projections—corticospinal and callosal projection neurons—develop from the same progenitors, but acquire strikingly different fates. The broad evolutionary conservation of known genes controlling cortical projection neuron fates raises the question of how the more narrowly conserved corticospinal and callosal projections evolved. We identify a microRNA cluster selectively expressed by corticospinal projection neurons and exclusive to placental mammals. One of these microRNAs promotes corticospinal fate via regulation of the callosal gene LMO4, suggesting a mechanism whereby microRNA regulation during development promotes evolution of neuronal diversity. The corticospinal tract is unique to mammals and the corpus callosum is unique to placental mammals (eutherians). The emergence of these structures is thought to underpin the evolutionary acquisition of complex motor and cognitive skills. Corticospinal motor neurons (CSMN) and callosal projection neurons (CPN) are the archetypal projection neurons of the corticospinal tract and corpus callosum, respectively. Although a number of conserved transcriptional regulators of CSMN and CPN development have been identified in vertebrates, none are unique to mammals and most are coexpressed across multiple projection neuron subtypes. Here, we discover 17 CSMN-enriched microRNAs (miRNAs), 15 of which map to a single genomic cluster that is exclusive to eutherians. One of these, miR-409-3p, promotes CSMN subtype identity in part via repression of LMO4, a key transcriptional regulator of CPN development. In vivo, miR-409-3p is sufficient to convert deep-layer CPN into CSMN. This is a demonstration of an evolutionarily acquired miRNA in eutherians that refines cortical projection neuron subtype development. Our findings implicate miRNAs in the eutherians’ increase in neuronal subtype and projection diversity, the anatomic underpinnings of their complex behavior.
Collapse
|
45
|
Transcription factor expression defines subclasses of developing projection neurons highly similar to single-cell RNA-seq subtypes. Proc Natl Acad Sci U S A 2020; 117:25074-25084. [PMID: 32948690 DOI: 10.1073/pnas.2008013117] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
We are only just beginning to catalog the vast diversity of cell types in the cerebral cortex. Such categorization is a first step toward understanding how diversification relates to function. All cortical projection neurons arise from a uniform pool of progenitor cells that lines the ventricles of the forebrain. It is still unclear how these progenitor cells generate the more than 50 unique types of mature cortical projection neurons defined by their distinct gene-expression profiles. Moreover, exactly how and when neurons diversify their function during development is unknown. Here we relate gene expression and chromatin accessibility of two subclasses of projection neurons with divergent morphological and functional features as they develop in the mouse brain between embryonic day 13 and postnatal day 5 in order to identify transcriptional networks that diversify neuron cell fate. We compare these gene-expression profiles with published profiles of single cells isolated from similar populations and establish that layer-defined cell classes encompass cell subtypes and developmental trajectories identified using single-cell sequencing. Given the depth of our sequencing, we identify groups of transcription factors with particularly dense subclass-specific regulation and subclass-enriched transcription factor binding motifs. We also describe transcription factor-adjacent long noncoding RNAs that define each subclass and validate the function of Myt1l in balancing the ratio of the two subclasses in vitro. Our multidimensional approach supports an evolving model of progressive restriction of cell fate competence through inherited transcriptional identities.
Collapse
|
46
|
Lipiec MA, Bem J, Koziński K, Chakraborty C, Urban-Ciećko J, Zajkowski T, Dąbrowski M, Szewczyk ŁM, Toval A, Ferran JL, Nagalski A, Wiśniewska MB. TCF7L2 regulates postmitotic differentiation programmes and excitability patterns in the thalamus. Development 2020; 147:dev.190181. [PMID: 32675279 PMCID: PMC7473649 DOI: 10.1242/dev.190181] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 07/08/2020] [Indexed: 12/14/2022]
Abstract
Neuronal phenotypes are controlled by terminal selector transcription factors in invertebrates, but only a few examples of such regulators have been provided in vertebrates. We hypothesised that TCF7L2 regulates different stages of postmitotic differentiation in the thalamus, and functions as a thalamic terminal selector. To investigate this hypothesis, we used complete and conditional knockouts of Tcf7l2 in mice. The connectivity and clustering of neurons were disrupted in the thalamo-habenular region in Tcf7l2-/- embryos. The expression of subregional thalamic and habenular transcription factors was lost and region-specific cell migration and axon guidance genes were downregulated. In mice with a postnatal Tcf7l2 knockout, the induction of genes that confer thalamic terminal electrophysiological features was impaired. Many of these genes proved to be direct targets of TCF7L2. The role of TCF7L2 in terminal selection was functionally confirmed by impaired firing modes in thalamic neurons in the mutant mice. These data corroborate the existence of master regulators in the vertebrate brain that control stage-specific genetic programmes and regional subroutines, maintain regional transcriptional network during embryonic development, and induce terminal selection postnatally.
Collapse
Affiliation(s)
- Marcin Andrzej Lipiec
- Centre of New Technologies, University of Warsaw, Banacha 2, 02-097 Warsaw, Poland.,Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096 Warsaw, Poland
| | - Joanna Bem
- Centre of New Technologies, University of Warsaw, Banacha 2, 02-097 Warsaw, Poland
| | - Kamil Koziński
- Centre of New Technologies, University of Warsaw, Banacha 2, 02-097 Warsaw, Poland
| | - Chaitali Chakraborty
- Centre of New Technologies, University of Warsaw, Banacha 2, 02-097 Warsaw, Poland
| | | | - Tomasz Zajkowski
- Centre of New Technologies, University of Warsaw, Banacha 2, 02-097 Warsaw, Poland
| | - Michał Dąbrowski
- Nencki Institute of Experimental Biology, Pasteur 3, 02-093 Warsaw, Poland
| | | | - Angel Toval
- Department of Human Anatomy and Psychobiology, School of Medicine, University of Murcia and IMIB-Arrixaca Institute, Campus de la Salud, 30120 El Palmar, Murcia, Spain
| | - José Luis Ferran
- Department of Human Anatomy and Psychobiology, School of Medicine, University of Murcia and IMIB-Arrixaca Institute, Campus de la Salud, 30120 El Palmar, Murcia, Spain
| | - Andrzej Nagalski
- Centre of New Technologies, University of Warsaw, Banacha 2, 02-097 Warsaw, Poland
| | | |
Collapse
|
47
|
Tomofuji Y, Takaba H, Suzuki HI, Benlaribi R, Martinez CDP, Abe Y, Morishita Y, Okamura T, Taguchi A, Kodama T, Takayanagi H. Chd4 choreographs self-antigen expression for central immune tolerance. Nat Immunol 2020; 21:892-901. [PMID: 32601470 DOI: 10.1038/s41590-020-0717-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 05/19/2020] [Indexed: 02/06/2023]
Abstract
Autoreactive T cells are eliminated in the thymus to prevent autoimmunity by promiscuous expression of tissue-restricted self-antigens in medullary thymic epithelial cells. This expression is dependent on the transcription factor Fezf2, as well as the transcriptional regulator Aire, but the entire picture of the transcriptional program has been obscure. Here, we found that the chromatin remodeler Chd4, also called Mi-2β, plays a key role in the self-antigen expression in medullary thymic epithelial cells. To maximize the diversity of self-antigen expression, Fezf2 and Aire utilized completely distinct transcriptional mechanisms, both of which were under the control of Chd4. Chd4 organized the promoter regions of Fezf2-dependent genes, while contributing to the Aire-mediated induction of self-antigens via super-enhancers. Mice deficient in Chd4 specifically in thymic epithelial cells exhibited autoimmune phenotypes, including T cell infiltration. Thus, Chd4 plays a critical role in integrating Fezf2- and Aire-mediated gene induction to establish central immune tolerance.
Collapse
Affiliation(s)
- Yoshihiko Tomofuji
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Takaba
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroshi I Suzuki
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Molecular Oncology, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Rayene Benlaribi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Cristian David Peña Martinez
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshihiro Abe
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasuyuki Morishita
- Department of Molecular Pathology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tadashi Okamura
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
- Section of Animal Models, National Center for Global Health and Medicine, Tokyo, Japan
| | - Akashi Taguchi
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Tatsuhiko Kodama
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
48
|
Kaur N, Han W, Li Z, Madrigal MP, Shim S, Pochareddy S, Gulden FO, Li M, Xu X, Xing X, Takeo Y, Li Z, Lu K, Imamura Kawasawa Y, Ballester-Lurbe B, Moreno-Bravo JA, Chédotal A, Terrado J, Pérez-Roger I, Koleske AJ, Sestan N. Neural Stem Cells Direct Axon Guidance via Their Radial Fiber Scaffold. Neuron 2020; 107:1197-1211.e9. [PMID: 32707082 DOI: 10.1016/j.neuron.2020.06.035] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 03/06/2020] [Accepted: 06/26/2020] [Indexed: 10/23/2022]
Abstract
Neural stem cells directly or indirectly generate all neurons and macroglial cells and guide migrating neurons by using a palisade-like scaffold made of their radial fibers. Here, we describe an unexpected role for the radial fiber scaffold in directing corticospinal and other axons at the junction between the striatum and globus pallidus. The maintenance of this scaffold, and consequently axon pathfinding, is dependent on the expression of an atypical RHO-GTPase, RND3/RHOE, together with its binding partner ARHGAP35/P190A, a RHO GTPase-activating protein, in the radial glia-like neural stem cells within the ventricular zone of the medial ganglionic eminence. This role is independent of RND3 and ARHGAP35 expression in corticospinal neurons, where they regulate dendritic spine formation, axon elongation, and pontine midline crossing in a FEZF2-dependent manner. The prevalence of neural stem cell scaffolds and their expression of RND3 and ARHGAP35 suggests that these observations might be broadly relevant for axon guidance and neural circuit formation.
Collapse
Affiliation(s)
- Navjot Kaur
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Wenqi Han
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Zhuo Li
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA; Graduate Program in Histology and Embryology, Zhengzhou University, 450001 Zhengzhou, China
| | - M Pilar Madrigal
- Department of Biomedical Sciences, School of Health Sciences and Veterinary School, Universidad Cardenal Herrera-CEU, CEU Universities, Moncada, 46113 Valencia, Spain
| | - Sungbo Shim
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA; Department of Biochemistry, Chungbuk National University, Cheongju 28644, Korea
| | - Sirisha Pochareddy
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Forrest O Gulden
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Mingfeng Li
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Xuming Xu
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Xiaojun Xing
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA; Yale Genome Editing Center, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Yutaka Takeo
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Zhen Li
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Kangrong Lu
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Yuka Imamura Kawasawa
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA; Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology and of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Begoña Ballester-Lurbe
- Department of Biomedical Sciences, School of Health Sciences and Veterinary School, Universidad Cardenal Herrera-CEU, CEU Universities, Moncada, 46113 Valencia, Spain
| | | | - Alain Chédotal
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - José Terrado
- Department of Biomedical Sciences, School of Health Sciences and Veterinary School, Universidad Cardenal Herrera-CEU, CEU Universities, Moncada, 46113 Valencia, Spain
| | - Ignacio Pérez-Roger
- Department of Biomedical Sciences, School of Health Sciences and Veterinary School, Universidad Cardenal Herrera-CEU, CEU Universities, Moncada, 46113 Valencia, Spain
| | - Anthony J Koleske
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA; Department of Molecular Biochemistry and Biophysics, Yale University, New Haven, CT 06520, USA
| | - Nenad Sestan
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA; Departments of Genetics, Psychiatry, and Comparative Medicine, Kavli Institute for Neuroscience, Program in Cellular Neuroscience, Neurodegeneration, and Repair, and Yale Child Study Center, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
49
|
Injured adult neurons regress to an embryonic transcriptional growth state. Nature 2020; 581:77-82. [PMID: 32376949 DOI: 10.1038/s41586-020-2200-5] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 02/13/2020] [Indexed: 12/25/2022]
Abstract
Grafts of spinal-cord-derived neural progenitor cells (NPCs) enable the robust regeneration of corticospinal axons and restore forelimb function after spinal cord injury1; however, the molecular mechanisms that underlie this regeneration are unknown. Here we perform translational profiling specifically of corticospinal tract (CST) motor neurons in mice, to identify their 'regenerative transcriptome' after spinal cord injury and NPC grafting. Notably, both injury alone and injury combined with NPC grafts elicit virtually identical early transcriptomic responses in host CST neurons. However, in mice with injury alone this regenerative transcriptome is downregulated after two weeks, whereas in NPC-grafted mice this transcriptome is sustained. The regenerative transcriptome represents a reversion to an embryonic transcriptional state of the CST neuron. The huntingtin gene (Htt) is a central hub in the regeneration transcriptome; deletion of Htt significantly attenuates regeneration, which shows that Htt has a key role in neural plasticity after injury.
Collapse
|
50
|
Remesal L, Roger-Baynat I, Chirivella L, Maicas M, Brocal-Ruiz R, Pérez-Villalba A, Cucarella C, Casado M, Flames N. PBX1 acts as terminal selector for olfactory bulb dopaminergic neurons. Development 2020; 147:dev.186841. [PMID: 32156753 DOI: 10.1242/dev.186841] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 02/24/2020] [Indexed: 02/03/2023]
Abstract
Neuronal specification is a protracted process that begins with the commitment of progenitor cells and culminates with the generation of mature neurons. Many transcription factors are continuously expressed during this process but it is presently unclear how these factors modify their targets as cells transition through different stages of specification. In olfactory bulb adult neurogenesis, the transcription factor PBX1 controls neurogenesis in progenitor cells and the survival of migrating neuroblasts. Here, we show that, at later differentiation stages, PBX1 also acts as a terminal selector for the dopaminergic neuron fate. PBX1 is also required for the morphological maturation of dopaminergic neurons and to repress alternative interneuron fates, findings that expand the known repertoire of terminal-selector actions. Finally, we reveal that the temporal diversification of PBX1 functions in neuronal specification is achieved, at least in part, through the dynamic regulation of alternative splicing. In Caenorhabditis elegans, PBX/CEH-20 also acts as a dopaminergic neuron terminal selector, which suggests an ancient role for PBX factors in the regulation of terminal differentiation of dopaminergic neurons.
Collapse
Affiliation(s)
- Laura Remesal
- Developmental Neurobiology Unit, Instituto de Biomedicina de Valencia, IBV-CSIC, 46010 Valencia, Spain
| | - Isabel Roger-Baynat
- Developmental Neurobiology Unit, Instituto de Biomedicina de Valencia, IBV-CSIC, 46010 Valencia, Spain
| | - Laura Chirivella
- Developmental Neurobiology Unit, Instituto de Biomedicina de Valencia, IBV-CSIC, 46010 Valencia, Spain
| | - Miren Maicas
- Developmental Neurobiology Unit, Instituto de Biomedicina de Valencia, IBV-CSIC, 46010 Valencia, Spain
| | - Rebeca Brocal-Ruiz
- Developmental Neurobiology Unit, Instituto de Biomedicina de Valencia, IBV-CSIC, 46010 Valencia, Spain
| | - Ana Pérez-Villalba
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BIOTECMED), and Departamento de Biología Celular, Biología Funcional y Antropología Física, Universidad de Valencia, 46100 Burjassot, Spain
| | - Carme Cucarella
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III (ISCIII), Metabolic Experimental Pathology Unit, Instituto de Biomedicina de Valencia, IBV-CSIC, 46010 Valencia, Spain
| | - Marta Casado
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III (ISCIII), Metabolic Experimental Pathology Unit, Instituto de Biomedicina de Valencia, IBV-CSIC, 46010 Valencia, Spain
| | - Nuria Flames
- Developmental Neurobiology Unit, Instituto de Biomedicina de Valencia, IBV-CSIC, 46010 Valencia, Spain
| |
Collapse
|