1
|
Saade M, Martí E. Early spinal cord development: from neural tube formation to neurogenesis. Nat Rev Neurosci 2025; 26:195-213. [PMID: 39915695 DOI: 10.1038/s41583-025-00906-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2025] [Indexed: 03/26/2025]
Abstract
As one of the simplest and most evolutionarily conserved parts of the vertebrate nervous system, the spinal cord serves as a key model for understanding the principles of nervous system construction. During embryonic development, the spinal cord originates from a population of bipotent stem cells termed neuromesodermal progenitors, which are organized within a transient embryonic structure known as the neural tube. Neural tube morphogenesis differs along its anterior-to-posterior axis: most of the neural tube (including the regions that will develop into the brain and the anterior spinal cord) forms via the bending and dorsal fusion of the neural groove, but the establishment of the posterior region of the neural tube involves de novo formation of a lumen within a solid medullary cord. The early spinal cord primordium consists of highly polarized neural progenitor cells organized into a pseudostratified epithelium. Tight regulation of the cell division modes of these progenitors drives the embryonic growth of the neural tube and initiates primary neurogenesis. A rich history of observational and functional studies across various vertebrate models has advanced our understanding of the cellular events underlying spinal cord development, and these foundational studies are beginning to inform our knowledge of human spinal cord development.
Collapse
Affiliation(s)
- Murielle Saade
- Department of Cells and Tissues, Instituto de Biología Molecular de Barcelona CSIC, Barcelona, Spain.
| | - Elisa Martí
- Department of Cells and Tissues, Instituto de Biología Molecular de Barcelona CSIC, Barcelona, Spain.
| |
Collapse
|
2
|
Lehr S, Brückner DB, Minchington TG, Greunz-Schindler M, Merrin J, Hannezo E, Kicheva A. Self-organized pattern formation in the developing mouse neural tube by a temporal relay of BMP signaling. Dev Cell 2025; 60:567-580.e14. [PMID: 39603235 DOI: 10.1016/j.devcel.2024.10.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 05/08/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024]
Abstract
Developing tissues interpret dynamic changes in morphogen activity to generate cell type diversity. To quantitatively study bone morphogenetic protein (BMP) signaling dynamics in the mouse neural tube, we developed an embryonic stem cell differentiation system tailored for growing tissues. Differentiating cells form striking self-organized patterns of dorsal neural tube cell types driven by sequential phases of BMP signaling that are observed both in vitro and in vivo. Data-driven biophysical modeling showed that these dynamics result from coupling fast negative feedback with slow positive regulation of signaling by the specification of an endogenous BMP source. Thus, in contrast to relays that propagate morphogen signaling in space, we identify a BMP signaling relay that operates in time. This mechanism allows for a rapid initial concentration-sensitive response that is robustly terminated, thereby regulating balanced sequential cell type generation. Our study provides an experimental and theoretical framework to understand how signaling dynamics are exploited in developing tissues.
Collapse
Affiliation(s)
- Stefanie Lehr
- Institute of Science and Technology Austria (ISTA), 3400 Klosterneuburg, Austria
| | - David B Brückner
- Institute of Science and Technology Austria (ISTA), 3400 Klosterneuburg, Austria
| | | | | | - Jack Merrin
- Institute of Science and Technology Austria (ISTA), 3400 Klosterneuburg, Austria
| | - Edouard Hannezo
- Institute of Science and Technology Austria (ISTA), 3400 Klosterneuburg, Austria.
| | - Anna Kicheva
- Institute of Science and Technology Austria (ISTA), 3400 Klosterneuburg, Austria.
| |
Collapse
|
3
|
Nandagopal S, Cha A, Jia BZ, Liao H, Comenho C, Lahav G, Wagner DE, Tsai TYC, Megason SG. Neural plate pre-patterning enables specification of intermediate neural progenitors in the spinal cord. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.09.632276. [PMID: 39829904 PMCID: PMC11741283 DOI: 10.1101/2025.01.09.632276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Dorsal-ventral patterning of neural progenitors in the posterior neural tube, which gives rise to the spinal cord, has served as a model system to understand how extracellular signals organize developing tissues. While previous work has shown that signaling gradients diversify progenitor fates at the dorsal and ventral ends of the tissue, the basis of fate specification in intermediate regions has remained unclear. Here we use zebrafish to investigate the neural plate, which precedes neural tube formation, and show that its pre-patterning by a distinct signaling environment enables intermediate fate specification. Systematic spatial analysis of transcription factor (TF) expression and signaling activity using a reference-based mapping approach shows that the neural plate is partitioned into a striking complexity of TF co-expression states that, in part, correspond to the activity of gastrulation signals such as FGF and Wnt that persist through axis extension. Using in toto analysis of cellular movement combined with fate mapping, we find that dbx1b-expressing intermediate progenitors (p0) originate from a neural-plate specific state characterized by transient co-expression of the TFs pax3a, olig4 and her3. Finally, we show that this state is defined by Wnt signaling in the posterior neural plate and that ectopic Wnt activation within pax3a/olig4+ cells is sufficient to promote dbx1b expression. Our data broadly support a model in which neural progenitor specification occurs through the sequential use of multiple signals to progressively diversify the neural tissue as it develops. This has implications for in vitro differentiation of spinal cord cell types and for understanding signal-based patterning in other developmental contexts.
Collapse
Affiliation(s)
- Sandy Nandagopal
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115
| | - Anna Cha
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115
| | - Bill Z. Jia
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115
| | - Hongyu Liao
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115
| | - Caroline Comenho
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115
| | - Galit Lahav
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115
| | - Daniel E. Wagner
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research and Department of Obstetrics, Gynecology and Reproductive Science, UCSF, San Francisco, CA 94143 USA
| | - Tony Y-C Tsai
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sean G. Megason
- Department of Systems Biology, Blavatnik Institute at Harvard Medical School, Boston, MA 02115
| |
Collapse
|
4
|
Yeung TJ, Wilkinson DG. Short-range Fgf signalling patterns hindbrain progenitors to induce the neurogenesis-to-oligodendrogenesis switch. Development 2024; 151:dev204256. [PMID: 39575980 DOI: 10.1242/dev.204256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 11/14/2024] [Indexed: 12/14/2024]
Abstract
In the vertebrate nervous system, neurogenesis generally precedes gliogenesis. The mechanisms driving the switch in cell type production and generation of the correct proportion of cell types remain unclear. Here, we show that Fgf20 signalling patterns progenitors to induce the switch from neurogenesis to oligodendrogenesis in the zebrafish hindbrain. Fgf20 emanating from earlier-born neurons signals at a short range to downregulate proneural gene expression in the segment centre with high spatial precision along both anterior-posterior and dorsal-ventral axes. This signal induces oligodendrocytes in the segment centre by upregulating olig2 and sox10 expression in pre-patterned competent progenitors. We show that the magnitude of proneural gene downregulation and the quantity of oligodendrocyte precursor cells specified is dependent on the extent of Fgf20 signalling. Overexpression of fgf20a induces precocious specification and differentiation of oligodendrocytes among olig2+ progenitors, resulting in an increase in oligodendrocytes at the expense of neurogenesis. Thus, Fgf20 signalling defines the proportion of each cell type produced. Taken together, Fgf20 signalling from earlier-born neurons patterns hindbrain segments spatially and temporally to induce the neurogenesis-to-oligodendrogenesis switch.
Collapse
Affiliation(s)
- Tim J Yeung
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | | |
Collapse
|
5
|
Marković A, Briscoe J, Page KM. Dynamics of positional information in the vertebrate neural tube. J R Soc Interface 2024; 21:20240414. [PMID: 39657793 PMCID: PMC11631457 DOI: 10.1098/rsif.2024.0414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/06/2024] [Accepted: 10/23/2024] [Indexed: 12/12/2024] Open
Abstract
In developing embryos, cells acquire distinct identities depending on their position in a tissue. Secreted signalling molecules, known as morphogens, act as long-range cues to provide the spatial information that controls these cell fate decisions. In several tissues, both the level and the duration of morphogen signalling appear to be important for determining cell fates. This is the case in the forming vertebrate nervous system where antiparallel morphogen gradients pattern the dorsal-ventral axis by partitioning the tissue into sharply delineated domains of molecularly distinct neural progenitors. How information in the gradients is decoded to generate precisely positioned boundaries of gene expression remains an open question. Here, we adopt tools from information theory to quantify the positional information in the neural tube and investigate how temporal changes in signalling could influence positional precision. The results reveal that the use of signalling dynamics, as well as the signalling level, substantially increases the precision possible for the estimation of position from morphogen gradients. This analysis links the dynamics of opposing morphogen gradients with precise pattern formation and provides an explanation for why time is used to impart positional information.
Collapse
Affiliation(s)
- Anđela Marković
- Department of Mathematics, University College London, LondonWC1E 6BT, UK
| | | | - Karen M. Page
- Department of Mathematics, University College London, LondonWC1E 6BT, UK
- Institute of Physics of Living Systems, University College London, London WC1E 6BT, UK
| |
Collapse
|
6
|
Manning E, Placzek M. Organizing activities of axial mesoderm. Curr Top Dev Biol 2024; 157:83-123. [PMID: 38556460 DOI: 10.1016/bs.ctdb.2024.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
For almost a century, developmental biologists have appreciated that the ability of the embryonic organizer to induce and pattern the body plan is intertwined with its differentiation into axial mesoderm. Despite this, we still have a relatively poor understanding of the contribution of axial mesoderm to induction and patterning of different body regions, and the manner in which axial mesoderm-derived information is interpreted in tissues of changing competence. Here, with a particular focus on the nervous system, we review the evidence that axial mesoderm notochord and prechordal mesoderm/mesendoderm act as organizers, discuss how their influence extends through the different axes of the developing organism, and describe how the ability of axial mesoderm to direct morphogenesis impacts on its role as a local organizer.
Collapse
Affiliation(s)
- Elizabeth Manning
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom; Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Marysia Placzek
- School of Biosciences, University of Sheffield, Sheffield, United Kingdom; Bateson Centre, University of Sheffield, Sheffield, United Kingdom; Neuroscience Institute, University of Sheffield, Sheffield, United Kingdom.
| |
Collapse
|
7
|
Teague S, Primavera G, Chen B, Liu ZY, Yao L, Freeburne E, Khan H, Jo K, Johnson C, Heemskerk I. Time-integrated BMP signaling determines fate in a stem cell model for early human development. Nat Commun 2024; 15:1471. [PMID: 38368368 PMCID: PMC10874454 DOI: 10.1038/s41467-024-45719-9] [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: 04/06/2023] [Accepted: 02/02/2024] [Indexed: 02/19/2024] Open
Abstract
How paracrine signals are interpreted to yield multiple cell fate decisions in a dynamic context during human development in vivo and in vitro remains poorly understood. Here we report an automated tracking method to follow signaling histories linked to cell fate in large numbers of human pluripotent stem cells (hPSCs). Using an unbiased statistical approach, we discover that measured BMP signaling history correlates strongly with fate in individual cells. We find that BMP response in hPSCs varies more strongly in the duration of signaling than the level. However, both the level and duration of signaling activity control cell fate choices only by changing the time integral. Therefore, signaling duration and level are interchangeable in this context. In a stem cell model for patterning of the human embryo, we show that signaling histories predict the fate pattern and that the integral model correctly predicts changes in cell fate domains when signaling is perturbed. Our data suggest that mechanistically, BMP signaling is integrated by SOX2.
Collapse
Affiliation(s)
- Seth Teague
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Gillian Primavera
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Bohan Chen
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Zong-Yuan Liu
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - LiAng Yao
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Emily Freeburne
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Hina Khan
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Kyoung Jo
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Craig Johnson
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Idse Heemskerk
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Center for Cell Plasticity and Organ Design, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Physics, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
8
|
Frith TJR, Briscoe J, Boezio GLM. From signalling to form: the coordination of neural tube patterning. Curr Top Dev Biol 2023; 159:168-231. [PMID: 38729676 DOI: 10.1016/bs.ctdb.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
The development of the vertebrate spinal cord involves the formation of the neural tube and the generation of multiple distinct cell types. The process starts during gastrulation, combining axial elongation with specification of neural cells and the formation of the neuroepithelium. Tissue movements produce the neural tube which is then exposed to signals that provide patterning information to neural progenitors. The intracellular response to these signals, via a gene regulatory network, governs the spatial and temporal differentiation of progenitors into specific cell types, facilitating the assembly of functional neuronal circuits. The interplay between the gene regulatory network, cell movement, and tissue mechanics generates the conserved neural tube pattern observed across species. In this review we offer an overview of the molecular and cellular processes governing the formation and patterning of the neural tube, highlighting how the remarkable complexity and precision of vertebrate nervous system arises. We argue that a multidisciplinary and multiscale understanding of the neural tube development, paired with the study of species-specific strategies, will be crucial to tackle the open questions.
Collapse
Affiliation(s)
| | - James Briscoe
- The Francis Crick Institute, London, United Kingdom.
| | | |
Collapse
|
9
|
Chinnaiya K, Placzek M. A Methodology for the Enzymatic Isolation of Embryonic Hypothalamus Tissue and Its Acute or Post-Culture Analysis by Multiplex Hybridisation Chain Reaction. Bio Protoc 2023; 13:e4898. [PMID: 38125731 PMCID: PMC10730952 DOI: 10.21769/bioprotoc.4898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/27/2023] [Accepted: 10/16/2023] [Indexed: 12/23/2023] Open
Abstract
The hypothalamus is an evolutionarily ancient part of the vertebrate ventral forebrain that integrates the dialogue between environment, peripheral body, and brain to centrally govern an array of physiologies and behaviours. Characterizing the mechanisms that control hypothalamic development illuminates both hypothalamic organization and function. Critical to the ability to unravel such mechanisms is the skill to isolate hypothalamic tissue, enabling both its acute analysis and its analysis after explant and culture. Tissue explants, in which cells develop in a manner analogous to their in vivo counterparts, are a highly effective tool to investigate the extrinsic signals and tissue-intrinsic self-organising features that drive hypothalamic development. The hypothalamus, however, is induced and patterned at neural tube stages of development, when the tissue is difficult to isolate, and its resident cells complex to define. No single molecular marker distinguishes early hypothalamic progenitor subsets from other cell types in the neural tube, and so their accurate dissection requires the simultaneous analysis of multiple proteins or mRNAs, techniques that were previously limited by antibody availability or were arduous to perform. Here, we overcome these challenges. We describe methodologies to precisely isolate early hypothalamic tissue from the embryonic chick at three distinct patterning stages and to culture hypothalamic explants in three-dimensional gels. We then describe optimised protocols for the analysis of embryos, isolated embryonic tissue, or cultured hypothalamic explants by multiplex hybridisation chain reaction. These methods can be applied to other vertebrates, including mouse, and to other tissue types. Key features • Detailed protocols for enzymatic isolation of embryonic chick hypothalamus at three patterning stages; methods can be extended to other vertebrates and tissues. • Brief methodologies for three-dimensional culture of hypothalamic tissue explants. • Optimised protocols for multiplex hybridisation chain reaction for analysis of embryos, isolated embryonic tissues, or explants.
Collapse
Affiliation(s)
| | - Marysia Placzek
- School of Biosciences, University of Sheffield, Sheffield, UK
| |
Collapse
|
10
|
Ho EK, Oatman HR, McFann SE, Yang L, Johnson HE, Shvartsman SY, Toettcher JE. Dynamics of an incoherent feedforward loop drive ERK-dependent pattern formation in the early Drosophila embryo. Development 2023; 150:dev201818. [PMID: 37602510 PMCID: PMC10482391 DOI: 10.1242/dev.201818] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 08/11/2023] [Indexed: 08/22/2023]
Abstract
Positional information in development often manifests as stripes of gene expression, but how stripes form remains incompletely understood. Here, we use optogenetics and live-cell biosensors to investigate the posterior brachyenteron (byn) stripe in early Drosophila embryos. This stripe depends on interpretation of an upstream ERK activity gradient and the expression of two target genes, tailless (tll) and huckebein (hkb), that exert antagonistic control over byn. We find that high or low doses of ERK signaling produce transient or sustained byn expression, respectively. Although tll transcription is always rapidly induced, hkb converts graded ERK inputs into a variable time delay. Nuclei thus interpret ERK amplitude through the relative timing of tll and hkb transcription. Antagonistic regulatory paths acting on different timescales are hallmarks of an incoherent feedforward loop, which is sufficient to explain byn dynamics and adds temporal complexity to the steady-state model of byn stripe formation. We further show that 'blurring' of an all-or-none stimulus through intracellular diffusion non-locally produces a byn stripe. Overall, we provide a blueprint for using optogenetics to dissect developmental signal interpretation in space and time.
Collapse
Affiliation(s)
- Emily K. Ho
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Harrison R. Oatman
- Program in Quantitative and Computational Biology, Princeton University, Princeton, NJ 08544, USA
| | - Sarah E. McFann
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544, USA
| | - Liu Yang
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Heath E. Johnson
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Stanislav Y. Shvartsman
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
- Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Center for Computational Biology, Flatiron Institute - Simons Foundation, New York, NY 10010, USA
| | - Jared E. Toettcher
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
11
|
Hettige NC, Fleming P, Semenak A, Zhang X, Peng H, Hagel MD, Théroux JF, Zhang Y, Ni A, Jefri M, Antonyan L, Alsuwaidi S, Schuppert A, Stumpf PS, Ernst C. FOXG1 targets BMP repressors and cell cycle inhibitors in human neural progenitor cells. Hum Mol Genet 2023; 32:2511-2522. [PMID: 37216650 PMCID: PMC10360395 DOI: 10.1093/hmg/ddad089] [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: 03/16/2023] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 05/24/2023] Open
Abstract
FOXG1 is a critical transcription factor in human brain where loss-of-function mutations cause a severe neurodevelopmental disorder, while increased FOXG1 expression is frequently observed in glioblastoma. FOXG1 is an inhibitor of cell patterning and an activator of cell proliferation in chordate model organisms but different mechanisms have been proposed as to how this occurs. To identify genomic targets of FOXG1 in human neural progenitor cells (NPCs), we engineered a cleavable reporter construct in endogenous FOXG1 and performed chromatin immunoprecipitation (ChIP) sequencing. We also performed deep RNA sequencing of NPCs from two females with loss-of-function mutations in FOXG1 and their healthy biological mothers. Integrative analyses of RNA and ChIP sequencing data showed that cell cycle regulation and Bone Morphogenic Protein (BMP) repression gene ontology categories were over-represented as FOXG1 targets. Using engineered brain cell lines, we show that FOXG1 specifically activates SMAD7 and represses CDKN1B. Activation of SMAD7 which inhibits BMP signaling may be one way that FOXG1 patterns the forebrain, while repression of cell cycle regulators such as CDKN1B may be one way that FOXG1 expands the NPC pool to ensure proper brain size. Our data reveal novel mechanisms on how FOXG1 may control forebrain patterning and cell proliferation in human brain development.
Collapse
Affiliation(s)
- Nuwan C Hettige
- Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada
- Psychiatric Genetics Group, Montreal, QC H4H 1R3, Canada
| | - Peter Fleming
- Psychiatric Genetics Group, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
| | - Amelia Semenak
- Psychiatric Genetics Group, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
| | - Xin Zhang
- Psychiatric Genetics Group, Montreal, QC H4H 1R3, Canada
| | - Huashan Peng
- Psychiatric Genetics Group, Montreal, QC H4H 1R3, Canada
| | - Marc-Daniel Hagel
- Joint Research Center for Computational Biomedicine, RWTH Aachen University, Aachen 52074, Germany
| | | | - Ying Zhang
- Psychiatric Genetics Group, Montreal, QC H4H 1R3, Canada
| | - Anjie Ni
- Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada
- Psychiatric Genetics Group, Montreal, QC H4H 1R3, Canada
| | - Malvin Jefri
- Psychiatric Genetics Group, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
| | - Lilit Antonyan
- Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada
- Psychiatric Genetics Group, Montreal, QC H4H 1R3, Canada
| | - Shaima Alsuwaidi
- Psychiatric Genetics Group, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
| | - Andreas Schuppert
- Joint Research Center for Computational Biomedicine, RWTH Aachen University, Aachen 52074, Germany
| | - Patrick S Stumpf
- Joint Research Center for Computational Biomedicine, RWTH Aachen University, Aachen 52074, Germany
| | - Carl Ernst
- Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada
- Psychiatric Genetics Group, Montreal, QC H4H 1R3, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
- Montreal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada
| |
Collapse
|
12
|
Teague S, Primavera G, Chen B, Freeburne E, Khan H, Jo K, Johnson C, Heemskerk I. The time integral of BMP signaling determines fate in a stem cell model for early human development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.10.536068. [PMID: 37090515 PMCID: PMC10120633 DOI: 10.1101/2023.04.10.536068] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
How paracrine signals are interpreted to yield multiple cell fate decisions in a dynamic context during human development in vivo and in vitro remains poorly understood. Here we report an automated tracking method to follow signaling histories linked to cell fate in large numbers of human pluripotent stem cells (hPSCs). Using an unbiased statistical approach, we discovered that measured BMP signaling history correlates strongly with fate in individual cells. We found that BMP response in hPSCs varies more strongly in the duration of signaling than the level. However, we discovered that both the level and duration of signaling activity control cell fate choices only by changing the time integral of signaling and that duration and level are therefore interchangeable in this context. In a stem cell model for patterning of the human embryo, we showed that signaling histories predict the fate pattern and that the integral model correctly predicts changes in cell fate domains when signaling is perturbed. Using an RNA-seq screen we then found that mechanistically, BMP signaling is integrated by SOX2.
Collapse
Affiliation(s)
- Seth Teague
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Gillian Primavera
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Bohan Chen
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan
| | - Emily Freeburne
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Hina Khan
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Kyoung Jo
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Craig Johnson
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Idse Heemskerk
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
- Center for Cell Plasticity and Organ Design, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Physics, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
13
|
Rekler D, Kalcheim C. Completion of neural crest cell production and emigration is regulated by retinoic-acid-dependent inhibition of BMP signaling. eLife 2022; 11:72723. [PMID: 35394423 PMCID: PMC8993216 DOI: 10.7554/elife.72723] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 03/02/2022] [Indexed: 12/21/2022] Open
Abstract
Production and emigration of neural crest cells is a transient process followed by the emergence of the definitive roof plate. The mechanisms regulating the end of neural crest ontogeny are poorly understood. Whereas early crest development is stimulated by mesoderm-derived retinoic acid, we report that the end of the neural crest period is regulated by retinoic acid synthesized in the dorsal neural tube. Inhibition of retinoic acid signaling in the neural tube prevents the normal upregulation of BMP inhibitors in the nascent roof plate and prolongs the period of BMP responsiveness which otherwise ceases close to roof plate establishment. Consequently, neural crest production and emigration are extended well into the roof plate stage. In turn, extending the activity of neural crest-specific genes inhibits the onset of retinoic acid synthesis in roof plate suggesting a mutual repressive interaction between neural crest and roof plate traits. Although several roof plate-specific genes are normally expressed in the absence of retinoic acid signaling, roof plate and crest markers are co-expressed in single cells and this domain also contains dorsal interneurons. Hence, the cellular and molecular architecture of the roof plate is compromised. Collectively, our results demonstrate that neural tube-derived retinoic acid, via inhibition of BMP signaling, is an essential factor responsible for the end of neural crest generation and the proper segregation of dorsal neural lineages. The division between the central nervous system – formed by the brain and spinal cord – and the peripheral nervous system – which consists of the neurons that sense and relay information to and from the body – takes place early during embryonic development. Initially, the nervous system consists of a tube of cells called the neural tube. From the top region of this tube, some cells change their shape, exit the tube and migrate to different places in the developing body. These cells are called the ‘neural crest’, and they form many different structures, including the peripheral nervous system. Neural crest cells keep leaving the neural tube for a period of time, but after that, the neural tube stops producing them. At this point, the region of the neural tube that had been producing neural crest cells becomes the ‘roof plate’ of the central nervous system, a structure that is essential for the development of specific groups of neurons in the brain and spinal cord. In bird embryos, a protein called bone morphogenetic protein (BMP) is essential for neural crest production because it triggers the migration of these cells away from the neural tube. Before the roof plate is formed, the activity of BMP is blocked by proteins known as BMP inhibitors, which stop more cells from leaving the neural tube. Around the time when neural crest formation stops, another molecule called retinoic acid begins to be synthesized in the top region of the neural tube. Rekler and Kalcheim asked whether retinoic acid is involved in the transition from neural crest to roof plate. To test this hypothesis, Rekler and Kalcheim blocked the activity of retinoic acid in the neural tube of quail embryos at the time when they should stop producing neural crest cells. This resulted in embryos in which the neural tube keeps producing neural crest cells after the roof plate has formed. In these embryos, individual cells in the resulting ‘roof plate’ produced both proteins that are normally only found in neural crest cells, and proteins typically exclusive to the roof plate. This suggests that, in the absence of retinoic acid activity, the segregation of neural crest identity from roof plate identity is compromised. Rekler and Kalcheim also found that, in the embryos where retinoic acid activity had been blocked, the cells in the area where the roof plate should be produced virtually no BMP inhibitors, and exhibited extended BMP activity. This allowed neural crest cells to continue forming and migrating away from the neural tube well after the period when they would stop in a normal embryo. These results indicate that retinoic acid stops the production of neural crest cells by repressing BMP activity in the roof plate of the neural tube. Rekler and Kalcheim’s experiments shed light on the mechanisms that allow the central and peripheral nervous systems to become segregated. This could increase our understanding of the origin of several neurodevelopmental disorders, potentially providing insights into their treatment or prevention. Additionally, the process of neural crest production and exit from the neural tube is highly similar to the process of metastasis in many invasive cancers. Thus, by understanding how the production of neural crest cells is terminated, it may be possible to learn how to prevent malignant cancer cells from spreading through the body.
Collapse
Affiliation(s)
- Dina Rekler
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
| | - Chaya Kalcheim
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, Israel
| |
Collapse
|
14
|
Vetter R, Iber D. Precision of morphogen gradients in neural tube development. Nat Commun 2022; 13:1145. [PMID: 35241686 PMCID: PMC8894346 DOI: 10.1038/s41467-022-28834-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 02/15/2022] [Indexed: 12/19/2022] Open
Abstract
Morphogen gradients encode positional information during development. How high patterning precision is achieved despite natural variation in both the morphogen gradients and in the readout process, is still largely elusive. Here, we show that the positional error of gradients in the mouse neural tube has previously been overestimated, and that the reported accuracy of the central progenitor domain boundaries in the mouse neural tube can be achieved with a single gradient, rather than requiring the simultaneous readout of opposing gradients. Consistently and independently, numerical simulations based on measured molecular noise levels likewise result in lower gradient variabilities than reported. Finally, we show that the patterning mechanism yields progenitor cell numbers with even greater precision than boundary positions, as gradient amplitude changes do not affect interior progenitor domain sizes. We conclude that single gradients can yield the observed developmental precision, which provides prospects for tissue engineering.
Collapse
Affiliation(s)
- Roman Vetter
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, 4058, Basel, Switzerland.
- Swiss Institute of Bioinformatics, Mattenstrasse 26, 4058, Basel, Switzerland.
| | - Dagmar Iber
- Department of Biosystems Science and Engineering, ETH Zürich, Mattenstrasse 26, 4058, Basel, Switzerland.
- Swiss Institute of Bioinformatics, Mattenstrasse 26, 4058, Basel, Switzerland.
| |
Collapse
|
15
|
Le Dréau G. BuMPing Into Neurogenesis: How the Canonical BMP Pathway Regulates Neural Stem Cell Divisions Throughout Space and Time. Front Neurosci 2022; 15:819990. [PMID: 35153664 PMCID: PMC8829030 DOI: 10.3389/fnins.2021.819990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 12/28/2021] [Indexed: 11/13/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are secreted factors that contribute to many aspects of the formation of the vertebrate central nervous system (CNS), from the initial shaping of the neural primordium to the maturation of the brain and spinal cord. In particular, the canonical (SMAD1/5/8-dependent) BMP pathway appears to play a key role during neurogenesis, its activity dictating neural stem cell fate decisions and thereby regulating the growth and homeostasis of the CNS. In this mini-review, I summarize accumulating evidence demonstrating how the canonical BMP activity promotes the amplification and/or maintenance of neural stem cells at different times and in diverse regions of the vertebrate CNS, and highlight findings suggesting that this function is evolutionarily conserved.
Collapse
|
16
|
Gupta S, Butler SJ. Getting in touch with your senses: Mechanisms specifying sensory interneurons in the dorsal spinal cord. WIREs Mech Dis 2021; 13:e1520. [PMID: 34730293 PMCID: PMC8459260 DOI: 10.1002/wsbm.1520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/10/2021] [Accepted: 01/11/2021] [Indexed: 11/18/2022]
Abstract
The spinal cord is functionally and anatomically divided into ventrally derived motor circuits and dorsally derived somatosensory circuits. Sensory stimuli originating either at the periphery of the body, or internally, are relayed to the dorsal spinal cord where they are processed by distinct classes of sensory dorsal interneurons (dIs). dIs convey sensory information, such as pain, heat or itch, either to the brain, and/or to the motor circuits to initiate the appropriate response. They also regulate the intensity of sensory information and are the major target for the opioid analgesics. While the developmental mechanisms directing ventral and dorsal cell fates have been hypothesized to be similar, more recent research has suggested that dI fates are specified by novel mechanisms. In this review, we will discuss the molecular events that specify dorsal neuronal patterning in the spinal cord, thereby generating diverse dI identities. We will then discuss how this molecular understanding has led to the development of robust stem cell methods to derive multiple spinal cell types, including the dIs, and the implication of these studies for treating spinal cord injuries and neurodegenerative diseases. This article is categorized under: Neurological Diseases > Stem Cells and Development.
Collapse
Affiliation(s)
- Sandeep Gupta
- Department of NeurobiologyUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| | - Samantha J. Butler
- Department of NeurobiologyUniversity of California, Los AngelesLos AngelesCaliforniaUSA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell ResearchUniversity of California, Los AngelesLos AngelesCaliforniaUSA
- Intellectual and Developmental Disabilities Research CenterUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| |
Collapse
|
17
|
Wang YF, Liu C, Xu PF. Deciphering and reconstitution of positional information in the human brain development. ACTA ACUST UNITED AC 2021; 10:29. [PMID: 34467458 PMCID: PMC8408296 DOI: 10.1186/s13619-021-00091-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/02/2021] [Indexed: 12/29/2022]
Abstract
Organoid has become a novel in vitro model to research human development and relevant disorders in recent years. With many improvements on the culture protocols, current brain organoids could self-organize into a complicated three-dimensional organization that mimics most of the features of the real human brain at the molecular, cellular, and further physiological level. However, lacking positional information, an important characteristic conveyed by gradients of signaling molecules called morphogens, leads to the deficiency of spatiotemporally regulated cell arrangements and cell–cell interactions in the brain organoid development. In this review, we will overview the role of morphogen both in the vertebrate neural development in vivo as well as the brain organoid culture in vitro, the strategies to apply morphogen concentration gradients in the organoid system and future perspectives of the brain organoid technology.
Collapse
Affiliation(s)
- Yi-Fan Wang
- Women's Hospital, and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Institute of Zhejiang University and University of Edinburgh, Jiaxing, Zhejiang, China.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Dr, Singapore, 117599, Singapore
| | - Cong Liu
- Women's Hospital, and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Peng-Fei Xu
- Women's Hospital, and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
18
|
Rekler D, Kalcheim C. From Neural Crest to Definitive Roof Plate: The Dynamic Behavior of the Dorsal Neural Tube. Int J Mol Sci 2021; 22:3911. [PMID: 33920095 PMCID: PMC8070085 DOI: 10.3390/ijms22083911] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 01/11/2023] Open
Abstract
Research on the development of the dorsal neural tube is particularly challenging. In this highly dynamic domain, a temporal transition occurs between early neural crest progenitors that undergo an epithelial-to-mesenchymal transition and exit the neural primordium, and the subsequent roof plate, a resident epithelial group of cells that constitutes the dorsal midline of the central nervous system. Among other functions, the roof plate behaves as an organizing center for the generation of dorsal interneurons. Despite extensive knowledge of the formation, emigration and migration of neural crest progenitors, little is known about the mechanisms leading to the end of neural crest production and the transition into a roof plate stage. Are these two mutually dependent or autonomously regulated processes? Is the generation of roof plate and dorsal interneurons induced by neural tube-derived factors throughout both crest and roof plate stages, respectively, or are there differences in signaling properties and responsiveness as a function of time? In this review, we discuss distinctive characteristics of each population and possible mechanisms leading to the shift between the above cell types.
Collapse
Affiliation(s)
| | - Chaya Kalcheim
- Department of Medical Neurobiology, Institute of Medical Research Israel-Canada (IMRIC) and the Edmond and Lily Safra Center for Brain Sciences (ELSC), Hebrew University of Jerusalem-Hadassah Medical School, P.O.Box 12272, Jerusalem 9112102, Israel;
| |
Collapse
|
19
|
Greenfeld H, Lin J, Mullins MC. The BMP signaling gradient is interpreted through concentration thresholds in dorsal-ventral axial patterning. PLoS Biol 2021; 19:e3001059. [PMID: 33481775 PMCID: PMC7857602 DOI: 10.1371/journal.pbio.3001059] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/03/2021] [Accepted: 01/07/2021] [Indexed: 12/24/2022] Open
Abstract
Bone Morphogenetic Protein (BMP) patterns the dorsal–ventral (DV) embryonic axis in all vertebrates, but it is unknown how cells along the DV axis interpret and translate the gradient of BMP signaling into differential gene activation that will give rise to distinct cell fates. To determine the mechanism of BMP morphogen interpretation in the zebrafish gastrula, we identified 57 genes that are directly activated by BMP signaling. By using Seurat analysis of single-cell RNA sequencing (scRNA-seq) data, we found that these genes are expressed in at least 3 distinct DV domains of the embryo. We distinguished between 3 models of BMP signal interpretation in which cells activate distinct gene expression through interpretation of thresholds of (1) the BMP signaling gradient slope; (2) the BMP signal duration; or (3) the level of BMP signal activation. We tested these 3 models using quantitative measurements of phosphorylated Smad5 (pSmad5) and by examining the spatial relationship between BMP signaling and activation of different target genes at single-cell resolution across the embryo. We found that BMP signaling gradient slope or BMP exposure duration did not account for the differential target gene expression domains. Instead, we show that cells respond to 3 distinct levels of BMP signaling activity to activate and position target gene expression. Together, we demonstrate that distinct pSmad5 threshold levels activate spatially distinct target genes to pattern the DV axis. This study tested three models of how a BMP morphogen gradient is translated into differential gene activation that specifies distinct cell fates, finding that BMP signal concentration thresholds, not gradient shape or signal duration, position three distinct gene activation domains.
Collapse
Affiliation(s)
- Hannah Greenfeld
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States of America
| | - Jerome Lin
- Institute for Biomedical Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States of America
| | - Mary C. Mullins
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States of America
- * E-mail:
| |
Collapse
|
20
|
Sasai N, Kadoya M, Ong Lee Chen A. Neural induction: Historical views and application to pluripotent stem cells. Dev Growth Differ 2021; 63:26-37. [PMID: 33289091 DOI: 10.1111/dgd.12703] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/22/2020] [Accepted: 11/02/2020] [Indexed: 12/20/2022]
Abstract
Embryonic stem (ES) cells are a useful experimental material to recapitulate the differentiation steps of early embryos, which are usually invisible and inaccessible from outside of the body, especially in mammals. ES cells have greatly facilitated the analyses of gene expression profiles and cell characteristics. In addition, understanding the mechanisms during neural differentiation is important for clinical purposes, such as developing new therapeutic methods or regenerative medicine. As neurons have very limited regenerative ability, neurodegenerative diseases are usually intractable, and patients suffer from the disease throughout their lifetimes. The functional cells generated from ES cells in vitro could replace degenerative areas by transplantation. In this review, we will first demonstrate the historical views and widely accepted concepts regarding the molecular mechanisms of neural induction and positional information to produce the specific types of neurons in model animals. Next, we will describe how these concepts have recently been applied to the research in the establishment of the methodology of neural differentiation from mammalian ES cells. Finally, we will focus on examples of the applications of differentiation systems to clinical purposes. Overall, the discussion will focus on how historical developmental studies are applied to state-of-the-art stem cell research.
Collapse
Affiliation(s)
- Noriaki Sasai
- Developmental Biomedical Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Minori Kadoya
- Developmental Biomedical Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Agnes Ong Lee Chen
- Developmental Biomedical Science, Nara Institute of Science and Technology, Ikoma, Japan
| |
Collapse
|
21
|
Alvarez S, Varadarajan SG, Butler SJ. Dorsal commissural axon guidance in the developing spinal cord. Curr Top Dev Biol 2020; 142:197-231. [PMID: 33706918 DOI: 10.1016/bs.ctdb.2020.10.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Commissural axons have been a key model system for identifying axon guidance signals in vertebrates. This review summarizes the current thinking about the molecular and cellular mechanisms that establish a specific commissural neural circuit: the dI1 neurons in the developing spinal cord. We assess the contribution of long- and short-range signaling while sequentially following the developmental timeline from the birth of dI1 neurons, to the extension of commissural axons first circumferentially and then contralaterally into the ventral funiculus.
Collapse
Affiliation(s)
- Sandy Alvarez
- Department of Neurobiology, University of California, Los Angeles, CA, United States; Molecular Biology Interdepartmental Doctoral Program, University of California, Los Angeles, CA, United States
| | | | - Samantha J Butler
- Department of Neurobiology, University of California, Los Angeles, CA, United States; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, United States.
| |
Collapse
|
22
|
Hart CG, Karimi-Abdolrezaee S. Bone morphogenetic proteins: New insights into their roles and mechanisms in CNS development, pathology and repair. Exp Neurol 2020; 334:113455. [PMID: 32877654 DOI: 10.1016/j.expneurol.2020.113455] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/18/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023]
Abstract
Bone morphogenetic proteins (BMPs) are a highly conserved and diverse family of proteins that play essential roles in various stages of development including the formation and patterning of the central nervous system (CNS). Bioavailability and function of BMPs are regulated by input from a plethora of transcription factors and signaling pathways. Intriguingly, recent literature has uncovered novel roles for BMPs in regulating homeostatic and pathological responses in the adult CNS. Basal levels of BMP ligands and receptors are widely expressed in the adult brain and spinal cord with differential expression patterns across CNS regions, cell types and subcellular locations. Recent evidence indicates that several BMP isoforms are transiently or chronically upregulated in the aged or pathological CNS. Genetic knockout and pharmacological studies have elucidated that BMPs regulate several aspects of CNS injury and repair including cell survival and differentiation, reactive astrogliosis and glial scar formation, axon regeneration, and myelin preservation and repair. Several BMP isoforms can be upregulated in the injured or diseased CNS simultaneously yet exert complementary or opposing effects on the endogenous cell responses after injury. Emerging studies also show that dysregulation of BMPs is associated with various CNS pathologies. Interestingly, modulation of BMPs can lead to beneficial or detrimental effects on CNS injury and repair mechanisms in a ligand, temporally or spatially specific manner, which reflect the complexity of BMP signaling. Given the significance of BMPs in neurodevelopment, a better understanding of their role in the context of injury may provide new therapeutic targets for the pathologic CNS. This review will provide a timely overview on the foundation and recent advancements in knowledge regarding the role and mechanisms of BMP signaling in the developing and adult CNS, and their implications in pathological responses and repair processes after injury or diseases.
Collapse
Affiliation(s)
- Christopher G Hart
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
23
|
Delás MJ, Briscoe J. Repressive interactions in gene regulatory networks: When you have no other choice. Curr Top Dev Biol 2020; 139:239-266. [PMID: 32450962 DOI: 10.1016/bs.ctdb.2020.03.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Tightly regulated gene expression programs, orchestrated by complex interactions between transcription factors, control cell type specification during development. Repressive interactions play a critical role in these networks, facilitating decision-making between two or more alternative cell fates. Here, we use the ventral neural tube as an example to illustrate how cross repressive interactions within a network drive pattern formation and specify cell types in response to a graded patterning signal. This and other systems serve to highlight how external signals are integrated through the cis regulatory elements controlling key genes and provide insight into the molecular underpinning of the process. Even the simplest networks can lead to counterintuitive results and we argue that a combination of experimental dissection and modeling approaches will be necessary to fully understand network behavior and the underlying design principles. Studying these gene regulatory networks as a whole ultimately allows us to extract fundamental properties applicable across systems that can expand our mechanistic understanding of how organisms develop.
Collapse
Affiliation(s)
| | - James Briscoe
- The Francis Crick Institute, London, United Kingdom.
| |
Collapse
|
24
|
Takebayashi-Suzuki K, Suzuki A. Intracellular Communication among Morphogen Signaling Pathways during Vertebrate Body Plan Formation. Genes (Basel) 2020; 11:E341. [PMID: 32213808 PMCID: PMC7141137 DOI: 10.3390/genes11030341] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 12/25/2022] Open
Abstract
During embryonic development in vertebrates, morphogens play an important role in cell fate determination and morphogenesis. Bone morphogenetic proteins (BMPs) belonging to the transforming growth factor-β (TGF-β) family control the dorsal-ventral (DV) patterning of embryos, whereas other morphogens such as fibroblast growth factor (FGF), Wnt family members, and retinoic acid (RA) regulate the formation of the anterior-posterior (AP) axis. Activation of morphogen signaling results in changes in the expression of target genes including transcription factors that direct cell fate along the body axes. To ensure the correct establishment of the body plan, the processes of DV and AP axis formation must be linked and coordinately regulated by a fine-tuning of morphogen signaling. In this review, we focus on the interplay of various intracellular regulatory mechanisms and discuss how communication among morphogen signaling pathways modulates body axis formation in vertebrate embryos.
Collapse
Affiliation(s)
- Kimiko Takebayashi-Suzuki
- Amphibian Research Center, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8526, Japan
| | - Atsushi Suzuki
- Graduate School of Integrated Sciences for Life, Amphibian Research Center, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8526, Japan
| |
Collapse
|
25
|
Sagner A, Briscoe J. Establishing neuronal diversity in the spinal cord: a time and a place. Development 2019; 146:146/22/dev182154. [DOI: 10.1242/dev.182154] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
ABSTRACT
The vertebrate spinal cord comprises multiple functionally distinct neuronal cell types arranged in characteristic positions. During development, these different types of neurons differentiate from transcriptionally distinct neural progenitors that are arrayed in discrete domains along the dorsal-ventral and anterior-posterior axes of the embryonic spinal cord. This organization arises in response to morphogen gradients acting upstream of a gene regulatory network, the architecture of which determines the spatial and temporal pattern of gene expression. In recent years, substantial progress has been made in deciphering the regulatory network that underlies the specification of distinct progenitor and neuronal cell identities. In this Review, we outline how distinct neuronal cell identities are established in response to spatial and temporal patterning systems, and outline novel experimental approaches to study the emergence and function of neuronal diversity in the spinal cord.
Collapse
|
26
|
Duval N, Vaslin C, Barata TC, Frarma Y, Contremoulins V, Baudin X, Nedelec S, Ribes VC. BMP4 patterns Smad activity and generates stereotyped cell fate organization in spinal organoids. Development 2019; 146:dev.175430. [PMID: 31239243 DOI: 10.1242/dev.175430] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 06/18/2019] [Indexed: 12/30/2022]
Abstract
Bone morphogenetic proteins (BMPs) are secreted regulators of cell fate in several developing tissues. In the embryonic spinal cord, they control the emergence of the neural crest, roof plate and distinct subsets of dorsal interneurons. Although a gradient of BMP activity has been proposed to determine cell type identity in vivo, whether this is sufficient for pattern formation in vitro is unclear. Here, we demonstrate that exposure to BMP4 initiates distinct spatial dynamics of BMP signalling within the self-emerging epithelia of both mouse and human pluripotent stem cell-derived spinal organoids. The pattern of BMP signalling results in the stereotyped spatial arrangement of dorsal neural tube cell types, and concentration, timing and duration of BMP4 exposure modulate these patterns. Moreover, differences in the duration of competence time-windows between mouse and human account for the species-specific tempo of neural differentiation. Together, this study describes efficient methods for generating patterned subsets of dorsal interneurons in spinal organoids and supports the conclusion that graded BMP activity orchestrates the spatial organization of the dorsal neural tube cellular diversity in mouse and human.
Collapse
Affiliation(s)
- Nathalie Duval
- Institut Jacques Monod, CNRS UMR7592, Université Paris Diderot, Sorbonne Paris Cité, 75205 Paris Cedex, France.,Institut Pasteur, Department of Developmental and Stem Cell Biology, CNRS URA 2578, 75015 Paris, France
| | - Célia Vaslin
- Inserm, UMR-S 1270, 75005 Paris, France.,Sorbonne Université, Science and Engineering Faculty, 75005 Paris, France.,Institut du Fer à Moulin, 75005 Paris, France
| | - Tiago C Barata
- Institut Jacques Monod, CNRS UMR7592, Université Paris Diderot, Sorbonne Paris Cité, 75205 Paris Cedex, France
| | - Youcef Frarma
- Institut Jacques Monod, CNRS UMR7592, Université Paris Diderot, Sorbonne Paris Cité, 75205 Paris Cedex, France
| | - Vincent Contremoulins
- Institut Jacques Monod, CNRS UMR7592, Université Paris Diderot, Sorbonne Paris Cité, 75205 Paris Cedex, France
| | - Xavier Baudin
- Institut Jacques Monod, CNRS UMR7592, Université Paris Diderot, Sorbonne Paris Cité, 75205 Paris Cedex, France.,ImagoSeine core facility of Institut Jacques Monod and member of France-BioImaging
| | - Stéphane Nedelec
- Inserm, UMR-S 1270, 75005 Paris, France .,Sorbonne Université, Science and Engineering Faculty, 75005 Paris, France.,Institut du Fer à Moulin, 75005 Paris, France
| | - Vanessa C Ribes
- Institut Jacques Monod, CNRS UMR7592, Université Paris Diderot, Sorbonne Paris Cité, 75205 Paris Cedex, France
| |
Collapse
|
27
|
Leung B, Shimeld SM. Evolution of vertebrate spinal cord patterning. Dev Dyn 2019; 248:1028-1043. [PMID: 31291046 DOI: 10.1002/dvdy.77] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/14/2019] [Accepted: 06/15/2019] [Indexed: 12/17/2022] Open
Abstract
The vertebrate spinal cord is organized across three developmental axes, anterior-posterior (AP), dorsal-ventral (DV), and medial-lateral (ML). Patterning of these axes is regulated by canonical intercellular signaling pathways: the AP axis by Wnt, fibroblast growth factor, and retinoic acid (RA), the DV axis by Hedgehog, Tgfβ, and Wnt, and the ML axis where proliferation is controlled by Notch. Developmental time plays an important role in which signal does what and when. Patterning across the three axes is not independent, but linked by interactions between signaling pathway components and their transcriptional targets. Combined this builds a sophisticated organ with many different types of cell in specific AP, DV, and ML positions. Two living lineages share phylum Chordata with vertebrates, amphioxus, and tunicates, while the jawless fish such as lampreys, survive as the most basally divergent vertebrate lineage. Genes and mechanisms shared between lampreys and other vertebrates tell us what predated vertebrates, while those also shared with other chordates tell us what evolved early in chordate evolution. Between these lie vertebrate innovations: genetic and developmental changes linked to evolution of new morphology. These include gene duplications, differences in how signals are received, and new regulatory connections between signaling pathways and their target genes.
Collapse
Affiliation(s)
- Brigid Leung
- Department of Zoology, University of Oxford, Oxford, UK
| | | |
Collapse
|
28
|
Kumar D, Nitzan E, Kalcheim C. YAP promotes neural crest emigration through interactions with BMP and Wnt activities. Cell Commun Signal 2019; 17:69. [PMID: 31228951 PMCID: PMC6589182 DOI: 10.1186/s12964-019-0383-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 06/04/2019] [Indexed: 12/12/2022] Open
Abstract
Background Premigratory neural crest progenitors undergo an epithelial-to-mesenchymal transition and leave the neural tube as motile cells. Previously, we showed that BMP generates trunk neural crest emigration through canonical Wnt signaling which in turn stimulates G1/S transition. The molecular network underlying this process is, however, not yet completely deciphered. Yes-associated-protein (YAP), an effector of the Hippo pathway, controls various aspects of development including cell proliferation, migration, survival and differentiation. In this study, we examined the possible involvement of YAP in neural crest emigration and its relationship with BMP and Wnt. Methods We implemented avian embryos in which levels of YAP gene activity were either reduced or upregulated by in ovo plasmid electroporation, and monitored effects on neural crest emigration, survival and proliferation. Neural crest-derived sensory neuron and melanocyte development were assessed upon gain of YAP function. Imunohistochemistry was used to assess YAP expression. In addition, the activity of specific signaling pathways including YAP, BMP and Wnt was monitored with specific reporters. Results We find that the Hippo pathway transcriptional co-activator YAP is expressed and is active in premigratory crest of avian embryos. Gain of YAP function stimulates neural crest emigration in vivo, and attenuating YAP inhibits cell exit. This is associated with an accumulation of FoxD3-expressing cells in the dorsal neural tube, with reduced proliferation, and enhanced apoptosis. Furthermore, gain of YAP function inhibits differentiation of Islet-1-positive sensory neurons and augments the number of EdnrB2-positive melanocytes. Using specific in vivo reporters, we show that loss of YAP function in the dorsal neural tube inhibits BMP and Wnt activities whereas gain of YAP function stimulates these pathways. Reciprocally, inhibition of BMP and Wnt signaling by noggin or Xdd1, respectively, downregulates YAP activity. In addition, YAP-dependent stimulation of neural crest emigration is compromised upon inhibition of either BMP or Wnt activities. Together, our results suggest a positive bidirectional cross talk between these pathways. Conclusions Our data show that YAP is necessary for emigration of neural crest progenitors. In addition, they incorporate YAP signaling into a BMP/Wnt-dependent molecular network responsible for emigration of trunk-level neural crest.
Collapse
Affiliation(s)
- Deepak Kumar
- Department of Medical Neurobiology, IMRIC and ELSC, Hebrew University of Jerusalem-Hadassah Medical School, P.O. Box 12272, 91120, Jerusalem, Israel
| | - Erez Nitzan
- Department of Medical Neurobiology, IMRIC and ELSC, Hebrew University of Jerusalem-Hadassah Medical School, P.O. Box 12272, 91120, Jerusalem, Israel
| | - Chaya Kalcheim
- Department of Medical Neurobiology, IMRIC and ELSC, Hebrew University of Jerusalem-Hadassah Medical School, P.O. Box 12272, 91120, Jerusalem, Israel.
| |
Collapse
|
29
|
A Notch-mediated, temporal asymmetry in BMP pathway activation promotes photoreceptor subtype diversification. PLoS Biol 2019; 17:e2006250. [PMID: 30703098 PMCID: PMC6372210 DOI: 10.1371/journal.pbio.2006250] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 02/12/2019] [Accepted: 01/22/2019] [Indexed: 11/19/2022] Open
Abstract
Neural progenitors produce neurons whose identities can vary as a function of the time that specification occurs. Here, we describe the heterochronic specification of two photoreceptor (PhR) subtypes in the zebrafish pineal gland. We find that accelerating PhR specification by impairing Notch signaling favors the early fate at the expense of the later fate. Using in vivo lineage tracing, we show that most pineal PhRs are born from a fate-restricted progenitor. Furthermore, sister cells derived from the division of PhR-restricted progenitors activate the bone morphogenetic protein (BMP) signaling pathway at different times after division, and this heterochrony requires Notch activity. Finally, we demonstrate that PhR identity is established as a function of when the BMP pathway is activated. We propose a novel model in which division of a progenitor with restricted potential generates sister cells with distinct identities via a temporal asymmetry in the activation of a signaling pathway. A major goal in the field of developmental neurobiology is to identify the mechanisms that underly the diversification of the subtypes of neurons that are needed for the function of the nervous system. When investigating these mechanisms, time is an often-overlooked variable. Here, we show that in the zebrafish pineal gland—a neuroendocrine organ containing mostly photoreceptors (PhRs) and projection neurons—different classes of PhRs appear in a temporal sequence. In this simple system, the decision to adopt a PhR fate is driven by the activation of the bone morphogenetic protein (BMP) signaling pathway. Following the final cell division of a PhR progenitor, the sister cells normally activate the BMP pathway at different times. When Notch signaling activity is abrogated, activation of the BMP pathway occurs earlier and synchronously, which in turn favors the development of early PhR fates at the expense of later fates. We propose a model in which preventing sister cells from activating a signaling pathway in a synchronous fashion after their final division allows diversification of cell fates.
Collapse
|
30
|
Andrews MG, Kong J, Novitch BG, Butler SJ. New perspectives on the mechanisms establishing the dorsal-ventral axis of the spinal cord. Curr Top Dev Biol 2018; 132:417-450. [PMID: 30797516 DOI: 10.1016/bs.ctdb.2018.12.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Distinct classes of neurons arise at different positions along the dorsal-ventral axis of the spinal cord leading to spinal neurons being segregated along this axis according to their physiological properties and functions. Thus, the neurons associated with motor control are generally located in, or adjacent to, the ventral horn whereas the interneurons (INs) that mediate sensory activities are present within the dorsal horn. Here, we review classic and recent studies examining the developmental mechanisms that establish the dorsal-ventral axis in the embryonic spinal cord. Intriguingly, while the cellular organization of the dorsal and ventral halves of the spinal cord looks superficially similar during early development, the underlying molecular mechanisms that establish dorsal vs ventral patterning are markedly distinct. For example, the ventral spinal cord is patterned by the actions of a single growth factor, sonic hedgehog (Shh) acting as a morphogen, i.e., concentration-dependent signal. Recent studies have shed light on the mechanisms by which the spatial and temporal gradient of Shh is transduced by cells to elicit the generation of different classes of ventral INs, and motor neurons (MNs). In contrast, the dorsal spinal cord is patterned by the action of multiple factors, most notably by members of the bone morphogenetic protein (BMP) and Wnt families. While less is known about dorsal patterning, recent studies have suggested that the BMPs do not act as morphogens to specify dorsal IN identities as previously proposed, rather each BMP has signal-specific activities. Finally, we consider the promise that elucidation of these mechanisms holds for neural repair.
Collapse
Affiliation(s)
- Madeline G Andrews
- Department of Neurobiology, University of California, Los Angeles, CA, United States; Neuroscience Graduate Program, University of California, Los Angeles, CA, United States
| | - Jennifer Kong
- Department of Neurobiology, University of California, Los Angeles, CA, United States; Neuroscience Graduate Program, University of California, Los Angeles, CA, United States
| | - Bennett G Novitch
- Department of Neurobiology, University of California, Los Angeles, CA, United States; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, United States
| | - Samantha J Butler
- Department of Neurobiology, University of California, Los Angeles, CA, United States; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, United States.
| |
Collapse
|
31
|
Yang Z, Jia H, Bai Y, Wang W. Bone morphogenetic protein 4 expression in the developing lumbosacral spinal cord of rat embryos with anorectal malformations. Int J Dev Neurosci 2018; 69:32-38. [PMID: 29959980 DOI: 10.1016/j.ijdevneu.2018.06.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 06/22/2018] [Accepted: 06/22/2018] [Indexed: 01/12/2023] Open
Abstract
Although there are improvements in treatment of anorectal malformations (ARMs), patients can still develop fecal incontinence, constipation, and soiling with loss in quality of life. Recent evidence suggests that malformations in the lumbosacral spinal cord are one of the factors that affect postoperative anorectal function. However, the underlying mechanism that produces these malformations has yet to be elucidated. The bone morphogenetic proteins (BMPs) comprise a large group of highly conserved molecules that are involved in multiple processes and play important roles in the formation, development, and differentiation of the spinal cord. This study was designed to investigate the levels of BMP4 expression in the lumbosacral spinal cord in ARMs rat embryos induced by ethylenethiourea (ETU). Specifically, we assessed the association of BMP4 levels with the maldevelopment of the lumbosacral spinal cord and whether BMP4 acted through the canonical intracellular pathway in embryonic rats with ARMs. BMP4 expression was confirmed with immunohistochemical staining, RT-qPCR and western blot analyses of embryonic day (E) 16, E17, E19 and E21 embryos, moreover Smad1/5 and pSmad1/5 expression were confirmed with western blot analyses at peak time point of BMP4 expression. Our results reveal that BMP4 expression in the lumbosacral spinal cord of ARMs rat embryos is decreased at both the mRNA and protein levels and could decrease the phosphorylation of smad1/5, when compared with their expression levels in normal tissue. These results also suggest that reductions in BMP4 expression were possibly responsible for dysfunction of the lumbosacral spinal cord during late developmental stages in ARMs fetal rats. Taken together, we conclude a role for BMP4 in the pathogenesis of lumbosacral spinal cord maldevelopment in developing ARMs rats.
Collapse
Affiliation(s)
- Zhonghua Yang
- Department of Pediatric Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Huimin Jia
- Department of Pediatric Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yuzuo Bai
- Department of Pediatric Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Weilin Wang
- Department of Pediatric Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
32
|
Andrews MG, Del Castillo LM, Ochoa-Bolton E, Yamauchi K, Smogorzewski J, Butler SJ. BMPs direct sensory interneuron identity in the developing spinal cord using signal-specific not morphogenic activities. eLife 2017; 6. [PMID: 28925352 PMCID: PMC5605194 DOI: 10.7554/elife.30647] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 08/24/2017] [Indexed: 02/06/2023] Open
Abstract
The Bone Morphogenetic Protein (BMP) family reiteratively signals to direct disparate cellular fates throughout embryogenesis. In the developing dorsal spinal cord, multiple BMPs are required to specify sensory interneurons (INs). Previous studies suggested that the BMPs act as concentration-dependent morphogens to direct IN identity, analogous to the manner in which sonic hedgehog patterns the ventral spinal cord. However, it remains unresolved how multiple BMPs would cooperate to establish a unified morphogen gradient. Our studies support an alternative model: BMPs have signal-specific activities directing particular IN fates. Using chicken and mouse models, we show that the identity, not concentration, of the BMP ligand directs distinct dorsal identities. Individual BMPs promote progenitor patterning or neuronal differentiation by their activation of different type I BMP receptors and distinct modulations of the cell cycle. Together, this study shows that a 'mix and match' code of BMP signaling results in distinct classes of sensory INs.
Collapse
Affiliation(s)
- Madeline G Andrews
- Department of Neurobiology, University of California, Los Angeles, United States.,Neuroscience Graduate Program, University of California, Los Angeles, United States.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, United States
| | - Lorenzo M Del Castillo
- Department of Neurobiology, University of California, Los Angeles, United States.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, United States.,CIRM Bridges to Research Program, California State University, Northridge, United States
| | - Eliana Ochoa-Bolton
- Department of Neurobiology, University of California, Los Angeles, United States.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, United States.,CIRM Bridges to Research Program, California State University, Northridge, United States
| | - Ken Yamauchi
- Department of Neurobiology, University of California, Los Angeles, United States.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, United States
| | - Jan Smogorzewski
- Department of Dermatology, University of Southern California, California, United States
| | - Samantha J Butler
- Department of Neurobiology, University of California, Los Angeles, United States.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, United States
| |
Collapse
|
33
|
Lai HC, Seal RP, Johnson JE. Making sense out of spinal cord somatosensory development. Development 2017; 143:3434-3448. [PMID: 27702783 DOI: 10.1242/dev.139592] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The spinal cord integrates and relays somatosensory input, leading to complex motor responses. Research over the past couple of decades has identified transcription factor networks that function during development to define and instruct the generation of diverse neuronal populations within the spinal cord. A number of studies have now started to connect these developmentally defined populations with their roles in somatosensory circuits. Here, we review our current understanding of how neuronal diversity in the dorsal spinal cord is generated and we discuss the logic underlying how these neurons form the basis of somatosensory circuits.
Collapse
Affiliation(s)
- Helen C Lai
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rebecca P Seal
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| | - Jane E Johnson
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
34
|
Demers CJ, Soundararajan P, Chennampally P, Cox GA, Briscoe J, Collins SD, Smith RL. Development-on-chip: in vitro neural tube patterning with a microfluidic device. Development 2017; 143:1884-92. [PMID: 27246712 PMCID: PMC4920155 DOI: 10.1242/dev.126847] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 03/24/2016] [Indexed: 01/17/2023]
Abstract
Embryogenesis is a highly regulated process in which the precise spatial and temporal release of soluble cues directs differentiation of multipotent stem cells into discrete populations of specialized adult cell types. In the spinal cord, neural progenitor cells are directed to differentiate into adult neurons through the action of mediators released from nearby organizing centers, such as the floor plate and paraxial mesoderm. These signals combine to create spatiotemporal diffusional landscapes that precisely regulate the development of the central nervous system (CNS). Currently, in vivo and ex vivo studies of these signaling factors present some inherent ambiguity. In vitro methods are preferred for their enhanced experimental clarity but often lack the technical sophistication required for biological realism. In this article, we present a versatile microfluidic platform capable of mimicking the spatial and temporal chemical environments found in vivo during neural tube development. Simultaneous opposing and/or orthogonal gradients of developmental morphogens can be maintained, resulting in neural tube patterning analogous to that observed in vivo. Summary: A microfluidic device mimics the spatial and temporal environment of neural tube development in vivo and enables the correct spatial organization of neural tube formation from stem cells in vitro.
Collapse
Affiliation(s)
- Christopher J Demers
- Microinstruments and Systems Laboratory, University of Maine, Orono, ME 04469, USA Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, USA The Francis Crick Institute, Mill Hill Laboratory, London NW7 1AA, UK
| | | | | | - Gregory A Cox
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, USA The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - James Briscoe
- The Francis Crick Institute, Mill Hill Laboratory, London NW7 1AA, UK
| | - Scott D Collins
- Microinstruments and Systems Laboratory, University of Maine, Orono, ME 04469, USA Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, USA
| | - Rosemary L Smith
- Microinstruments and Systems Laboratory, University of Maine, Orono, ME 04469, USA Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, USA
| |
Collapse
|
35
|
Lemke KA, Aghayee A, Ashton RS. Deriving, regenerating, and engineering CNS tissues using human pluripotent stem cells. Curr Opin Biotechnol 2017; 47:36-42. [PMID: 28605638 DOI: 10.1016/j.copbio.2017.05.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 05/08/2017] [Indexed: 01/08/2023]
Abstract
Progress in deriving a spectrum of central nervous system cell phenotypes from human pluripotent stem cells has spurred significant advances in in vitro modeling and development of regenerative therapies for neurological disorders. While the clinical impact of these advances is still being evaluated, their integration with advanced tissue engineering methodologies and therapeutic approaches that induce neural circuit plasticity, respectively, remain underexplored frontiers.
Collapse
Affiliation(s)
- Kristen A Lemke
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53706, United States
| | - Alireza Aghayee
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53706, United States; Department of Material Science and Engineering, University of Wisconsin-Madison, Madison, WI 53715, United States
| | - Randolph S Ashton
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI 53706, United States; Department of Material Science and Engineering, University of Wisconsin-Madison, Madison, WI 53715, United States; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53715, United States.
| |
Collapse
|
36
|
Briscoe J, Kicheva A. The physics of development 100 years after D'Arcy Thompson's “On Growth and Form”. Mech Dev 2017; 145:26-31. [DOI: 10.1016/j.mod.2017.03.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/22/2017] [Accepted: 03/28/2017] [Indexed: 12/30/2022]
|
37
|
Kabayiza KU, Masgutova G, Harris A, Rucchin V, Jacob B, Clotman F. The Onecut Transcription Factors Regulate Differentiation and Distribution of Dorsal Interneurons during Spinal Cord Development. Front Mol Neurosci 2017; 10:157. [PMID: 28603487 PMCID: PMC5445119 DOI: 10.3389/fnmol.2017.00157] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 05/08/2017] [Indexed: 01/09/2023] Open
Abstract
During embryonic development, the dorsal spinal cord generates numerous interneuron populations eventually involved in motor circuits or in sensory networks that integrate and transmit sensory inputs from the periphery. The molecular mechanisms that regulate the specification of these multiple dorsal neuronal populations have been extensively characterized. In contrast, the factors that contribute to their diversification into smaller specialized subsets and those that control the specific distribution of each population in the developing spinal cord remain unknown. Here, we demonstrate that the Onecut transcription factors, namely Hepatocyte Nuclear Factor-6 (HNF-6) (or OC-1), OC-2 and OC-3, regulate the diversification and the distribution of spinal dorsal interneuron (dINs). Onecut proteins are dynamically and differentially distributed in spinal dINs during differentiation and migration. Analyzes of mutant embryos devoid of Onecut factors in the developing spinal cord evidenced a requirement in Onecut proteins for proper production of a specific subset of dI5 interneurons. In addition, the distribution of dI3, dI5 and dI6 interneuron populations was altered. Hence, Onecut transcription factors control genetic programs that contribute to the regulation of spinal dIN diversification and distribution during embryonic development.
Collapse
Affiliation(s)
- Karolina U Kabayiza
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Neural DifferentiationBrussels, Belgium.,Biology Department, School of Science, College of Science and Technology, University of RwandaButare, Rwanda
| | - Gauhar Masgutova
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Neural DifferentiationBrussels, Belgium
| | - Audrey Harris
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Neural DifferentiationBrussels, Belgium
| | - Vincent Rucchin
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Neural DifferentiationBrussels, Belgium
| | - Benvenuto Jacob
- Université catholique de Louvain, Institute of Neuroscience, System and Cognition DivisionBrussels, Belgium
| | - Frédéric Clotman
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Neural DifferentiationBrussels, Belgium
| |
Collapse
|
38
|
Taneyhill LA, Schiffmacher AT. Should I stay or should I go? Cadherin function and regulation in the neural crest. Genesis 2017; 55. [PMID: 28253541 DOI: 10.1002/dvg.23028] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 02/21/2017] [Accepted: 02/27/2017] [Indexed: 12/20/2022]
Abstract
Our increasing comprehension of neural crest cell development has reciprocally advanced our understanding of cadherin expression, regulation, and function. As a transient population of multipotent stem cells that significantly contribute to the vertebrate body plan, neural crest cells undergo a variety of transformative processes and exhibit many cellular behaviors, including epithelial-to-mesenchymal transition (EMT), motility, collective cell migration, and differentiation. Multiple studies have elucidated regulatory and mechanistic details of specific cadherins during neural crest cell development in a highly contextual manner. Collectively, these results reveal that gradual changes within neural crest cells are accompanied by often times subtle, yet important, alterations in cadherin expression and function. The primary focus of this review is to coalesce recent data on cadherins in neural crest cells, from their specification to their emergence as motile cells soon after EMT, and to highlight the complexities of cadherin expression beyond our current perceptions, including the hypothesis that the neural crest EMT is a transition involving a predominantly singular cadherin switch. Further advancements in genetic approaches and molecular techniques will provide greater opportunities to integrate data from various model systems in order to distinguish unique or overlapping functions of cadherins expressed at any point throughout the ontogeny of the neural crest.
Collapse
Affiliation(s)
- Lisa A Taneyhill
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, 20742
| | - Andrew T Schiffmacher
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, 20742
| |
Collapse
|
39
|
Sonic -'Jack-of-All-Trades' in Neural Circuit Formation. J Dev Biol 2017; 5:jdb5010002. [PMID: 29615560 PMCID: PMC5831768 DOI: 10.3390/jdb5010002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/22/2017] [Accepted: 02/01/2017] [Indexed: 12/23/2022] Open
Abstract
As reflected by the term morphogen, molecules such as Shh and Wnts were identified based on their role in early development when they instruct precursor cells to adopt a specific cell fate. Only much later were they implicated in neural circuit formation. Both in vitro and in vivo studies indicated that morphogens direct axons during their navigation through the developing nervous system. Today, the best understood role of Shh and Wnt in axon guidance is their effect on commissural axons in the spinal cord. Shh was shown to affect commissural axons both directly and indirectly via its effect on Wnt signaling. In fact, throughout neural circuit formation there is cross-talk and collaboration of Shh and Wnt signaling. Thus, although the focus of this review is on the role of Shh in neural circuit formation, a separation from Wnt signaling is not possible.
Collapse
|
40
|
Watanabe M, Fung ES, Chan FB, Wong JS, Coutts M, Monuki ES. BMP4 acts as a dorsal telencephalic morphogen in a mouse embryonic stem cell culture system. Biol Open 2016; 5:1834-1843. [PMID: 27815243 PMCID: PMC5200901 DOI: 10.1242/bio.012021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 10/28/2016] [Indexed: 12/25/2022] Open
Abstract
The concept of a morphogen - a molecule that specifies two or more cell fates in a concentration-dependent manner - is paradigmatic in developmental biology. Much remains unknown, however, about the existence of morphogens in the developing vertebrate central nervous system (CNS), including the mouse dorsal telencephalic midline (DTM). Bone morphogenetic proteins (BMPs) are candidate DTM morphogens, and our previous work demonstrated BMP4 sufficiency to induce one DTM cell fate - that of choroid plexus epithelial cells (CPECs) - in a mouse embryonic stem cell (mESC) culture system. Here we used BMP4 in a modified mESC culture system to derive a second DTM fate, the cortical hem (CH). CH and CPEC markers were induced by BMP4 in a concentration-dependent manner consistent with in vivo development. BMP4 concentrations that led to CH fate also promoted markers for Cajal-Retzius neurons, which are known CH derivatives. Interestingly, single BMP4 administrations also sufficed for appropriate temporal regulation of CH, CPEC, and cortical genes, with initially broad and overlapping dose-response profiles that sharpened over time. BMP4 concentrations that yielded CH- or CPEC-enriched populations also had different steady-state levels of phospho-SMAD1/5/8, suggesting that differences in BMP signaling intensity underlie DTM fate choice. Surprisingly, inactivation of the cortical selector gene Lhx2 did not affect DTM expression levels, dose-response profiles, or timing in response to BMP4, although neural progenitor genes were downregulated. These data indicate that BMP4 can act as a classic morphogen to orchestrate both spatial and temporal aspects of DTM fate acquisition, and can do so in the absence of Lhx2.
Collapse
Affiliation(s)
- Momoko Watanabe
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California Irvine, Irvine, CA 92697-2300, USA
| | - Ernest S Fung
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA 92697-4800, USA
| | - Felicia B Chan
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA 92697-4800, USA
| | - Jessica S Wong
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA 92697-4800, USA
| | - Margaret Coutts
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA 92697-4800, USA
| | - Edwin S Monuki
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California Irvine, Irvine, CA 92697-2300, USA
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA 92697-4800, USA
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, CA 92697-1705, USA
| |
Collapse
|
41
|
Kicheva A, Briscoe J. Developmental Pattern Formation in Phases. Trends Cell Biol 2016; 25:579-591. [PMID: 26410404 DOI: 10.1016/j.tcb.2015.07.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 06/12/2015] [Accepted: 07/17/2015] [Indexed: 01/20/2023]
Abstract
Cells in developing organs undergo a series of changes in their transcriptional state until a complete repertoire of cell types is specified. These changes in cell identity, together with the control of tissue growth, determine the pattern of gene expression in the tissue. Recent studies explore the dynamics of pattern formation during development and provide new insights into the control mechanisms. Changes in morphogen signalling and transcriptional networks control the specification of cell types. This is often followed by a distinct second phase, where pattern is elaborated by tissue growth. Here, we discuss the transitions between distinct phases in pattern formation. We consider the implications of the underlying mechanisms for understanding how reproducible patterns form during development.
Collapse
Affiliation(s)
- Anna Kicheva
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, NW71AA, UK.
| | - James Briscoe
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, NW71AA, UK.
| |
Collapse
|
42
|
Nitzan E, Avraham O, Kahane N, Ofek S, Kumar D, Kalcheim C. Dynamics of BMP and Hes1/Hairy1 signaling in the dorsal neural tube underlies the transition from neural crest to definitive roof plate. BMC Biol 2016; 14:23. [PMID: 27012662 PMCID: PMC4806459 DOI: 10.1186/s12915-016-0245-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/10/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The dorsal midline region of the neural tube that results from closure of the neural folds is generally termed the roof plate (RP). However, this domain is highly dynamic and complex, and is first transiently inhabited by prospective neural crest (NC) cells that sequentially emigrate from the neuroepithelium. It only later becomes the definitive RP, the dorsal midline cells of the spinal cord. We previously showed that at the trunk level of the axis, prospective RP progenitors originate ventral to the premigratory NC and progressively reach the dorsal midline following NC emigration. However, the molecular mechanisms underlying the end of NC production and formation of the definitive RP remain virtually unknown. RESULTS Based on distinctive cellular and molecular traits, we have defined an initial NC and a subsequent RP stage, allowing us to investigate the mechanisms responsible for the transition between the two phases. We demonstrate that in spite of the constant production of BMP4 in the dorsal tube at both stages, RP progenitors only transiently respond to the ligand and lose competence shortly before they arrive at their final location. In addition, exposure of dorsal tube cells at the NC stage to high levels of BMP signaling induces premature RP traits, such as Hes1/Hairy1, while concomitantly inhibiting NC production. Reciprocally, early inhibition of BMP signaling prevents Hairy1 mRNA expression at the RP stage altogether, suggesting that BMP is both necessary and sufficient for the development of this RP-specific trait. Furthermore, when Hes1/Hairy1 is misexpressed at the NC stage, it inhibits BMP signaling and downregulates BMPR1A/Alk3 mRNA expression, transcription of BMP targets such as Foxd3, cell-cycle progression, and NC emigration. Reciprocally, Foxd3 inhibits Hairy1, suggesting that repressive cross-interactions at the level of, and downstream from, BMP ensure the temporal separation between both lineages. CONCLUSIONS Together, our data suggest that BMP signaling is important both for NC and RP formation. Given that these two structures develop sequentially, we speculate that the longer exposure of RP progenitors to BMP compared with that of premigratory NC cells may be translated into a higher signaling level in the former. This induces changes in responsiveness to BMP, most likely by downregulating the expression of Alk3 receptors and, consequently, of BMP-dependent downstream transcription factors, which exhibit spatial complementary expression patterns and mutually repress each other to generate alternative fates. This molecular dynamic is likely to account for the transition between the NC and definitive RP stages and thus be responsible for the segregation between central and peripheral lineages during neural development.
Collapse
Affiliation(s)
- Erez Nitzan
- Department of Medical Neurobiology, IMRIC and ELSC, Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, 9112102,, PO Box 12272,, Israel.,Present Address: Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Oshri Avraham
- Department of Medical Neurobiology, IMRIC and ELSC, Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, 9112102,, PO Box 12272,, Israel.,Present address: Department of Genetics, Washington University, St. Louis, MO, USA
| | - Nitza Kahane
- Department of Medical Neurobiology, IMRIC and ELSC, Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, 9112102,, PO Box 12272,, Israel
| | - Shai Ofek
- Department of Medical Neurobiology, IMRIC and ELSC, Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, 9112102,, PO Box 12272,, Israel
| | - Deepak Kumar
- Department of Medical Neurobiology, IMRIC and ELSC, Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, 9112102,, PO Box 12272,, Israel
| | - Chaya Kalcheim
- Department of Medical Neurobiology, IMRIC and ELSC, Hebrew University of Jerusalem-Hadassah Medical School, Jerusalem, 9112102,, PO Box 12272,, Israel.
| |
Collapse
|
43
|
Luehders K, Sasai N, Davaapil H, Kurosawa-Yoshida M, Hiura H, Brah T, Ohnuma SI. The small leucine-rich repeat secreted protein Asporin induces eyes in Xenopus embryos through the IGF signalling pathway. Development 2016; 142:3351-61. [PMID: 26443635 DOI: 10.1242/dev.124438] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Small leucine-rich repeat proteoglycan (SLRP) family proteins play important roles in a number of biological events. Here, we demonstrate that the SLRP family member Asporin (ASPN) plays a crucial role in the early stages of eye development in Xenopus embryos. During embryogenesis, ASPN is broadly expressed in the neuroectoderm of the embryo. Overexpression of ASPN causes the induction of ectopic eyes. By contrast, blocking ASPN function with a morpholino oligonucleotide (ASPN-MO) inhibits eye formation, indicating that ASPN is an essential factor for eye development. Detailed molecular analyses revealed that ASPN interacts with insulin growth factor receptor (IGFR) and is essential for activating the IGF receptor-mediated intracellular signalling pathway. Moreover, ASPN perturbed the Wnt, BMP and Activin signalling pathways, suggesting that ASPN thereby creates a favourable environment in which the IGF signal can dominate. ASPN is thus a novel secreted molecule essential for eye induction through the coordination of multiple signalling pathways.
Collapse
Affiliation(s)
- Kristin Luehders
- Ocular Biology and Therapeutic unit (ORBIT), Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Noriaki Sasai
- Ocular Biology and Therapeutic unit (ORBIT), Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK Developmental Biomedical Science, Graduate School of Biological Sciences, Nara Institute of Science and Technology (NAIST), 8916-5, Takayama-cho, Ikoma 630-0192, Japan
| | - Hongorzul Davaapil
- Ocular Biology and Therapeutic unit (ORBIT), Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Maiko Kurosawa-Yoshida
- Department of Oncology, The Hutchison/MRC Research Centre, University of Cambridge, Hills Road, Cambridge CB2 2XZ, UK
| | - Hitoshi Hiura
- Ocular Biology and Therapeutic unit (ORBIT), Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Tara Brah
- Ocular Biology and Therapeutic unit (ORBIT), Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Shin-ichi Ohnuma
- Ocular Biology and Therapeutic unit (ORBIT), Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK Department of Oncology, The Hutchison/MRC Research Centre, University of Cambridge, Hills Road, Cambridge CB2 2XZ, UK
| |
Collapse
|
44
|
Ai Z, Xiang Z, Li Y, Liu G, Wang H, Zheng Y, Qiu X, Zhao S, Zhu X, Li Y, Ji W, Li T. Conversion of monkey fibroblasts to transplantable telencephalic neuroepithelial stem cells. Biomaterials 2016; 77:53-65. [DOI: 10.1016/j.biomaterials.2015.10.079] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 10/27/2015] [Accepted: 10/29/2015] [Indexed: 12/11/2022]
|
45
|
Sox6 suppression induces RA-dependent apoptosis mediated by BMP-4 expression during neuronal differentiation in P19 cells. Mol Cell Biochem 2015; 412:49-57. [PMID: 26590087 PMCID: PMC4718955 DOI: 10.1007/s11010-015-2607-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/14/2015] [Indexed: 11/01/2022]
Abstract
Sox6 is a transcription factor that induces neuronal differentiation in P19 cells; its suppression not only inhibits neuronal differentiation but also induces retinoic acid (RA)-dependent apoptosis of P19 cells. In the present study, we found that Sox6 suppression-induced apoptosis was mediated by activation of caspase 9 and 3. Moreover, we noted a weak leakage of cytochrome c into the cytoplasm from the mitochondria, indicating that apoptosis occurs through a mitochondrial pathway in Sox6-suppressed P19 (P19[anti-Sox6]) cells. Sox6 suppression in the presence of RA also induced the expression and secretion of bone morphogenetic protein 4 (BMP-4). Addition of an anti-BMP-4 antibody for neutralization increased cell viability and led to RA-dependent death of P19[anti-Sox6] cells. Our results indicate that Sox6 suppression induces RA-dependent cell death of P19 cells, mediated by BMP-4 expression and secretion. Normally, high Sox6 expression leads to RA-mediated neuronal differentiation in P19 cells; however, Sox6 deficiency induces production and secretion of BMP-4, which mediates selective cell death. Our findings suggest that Sox6 contributes to cell survival by suppressing BMP-4 transcription during neuronal differentiation.
Collapse
|
46
|
Johnson MD, Reeder JE, O’Connell M. Bone morphogenetic protein-4 and 7 and receptors regulate vascular endothelial growth factor and receptors in human fetal leptomeninges. Neurosci Lett 2015; 606:225-30. [DOI: 10.1016/j.neulet.2015.08.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 08/09/2015] [Accepted: 08/17/2015] [Indexed: 12/11/2022]
|
47
|
Corallo D, Trapani V, Bonaldo P. The notochord: structure and functions. Cell Mol Life Sci 2015; 72:2989-3008. [PMID: 25833128 PMCID: PMC11114051 DOI: 10.1007/s00018-015-1897-z] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 03/23/2015] [Accepted: 03/26/2015] [Indexed: 01/08/2023]
Abstract
The notochord is an embryonic midline structure common to all members of the phylum Chordata, providing both mechanical and signaling cues to the developing embryo. In vertebrates, the notochord arises from the dorsal organizer and it is critical for proper vertebrate development. This evolutionary conserved structure located at the developing midline defines the primitive axis of embryos and represents the structural element essential for locomotion. Besides its primary structural function, the notochord is also a source of developmental signals that patterns surrounding tissues. Among the signals secreted by the notochord, Hedgehog proteins play key roles during embryogenesis. The Hedgehog signaling pathway is a central regulator of embryonic development, controlling the patterning and proliferation of a wide variety of organs. In this review, we summarize the current knowledge on notochord structure and functions, with a particular emphasis on the key developmental events that take place in vertebrates. Moreover, we discuss some genetic studies highlighting the phenotypic consequences of impaired notochord development, which enabled to understand the molecular basis of different human congenital defects and diseases.
Collapse
Affiliation(s)
- Diana Corallo
- Department of Molecular Medicine, University of Padova, Viale G. Colombo 3, 35131 Padua, Italy
| | - Valeria Trapani
- Department of Molecular Medicine, University of Padova, Viale G. Colombo 3, 35131 Padua, Italy
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, Viale G. Colombo 3, 35131 Padua, Italy
| |
Collapse
|
48
|
Deterministic HOX patterning in human pluripotent stem cell-derived neuroectoderm. Stem Cell Reports 2015; 4:632-44. [PMID: 25843047 PMCID: PMC4400649 DOI: 10.1016/j.stemcr.2015.02.018] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 02/23/2015] [Accepted: 02/24/2015] [Indexed: 11/25/2022] Open
Abstract
Colinear HOX expression during hindbrain and spinal cord development diversifies and assigns regional neural phenotypes to discrete rhombomeric and vertebral domains. Despite the precision of HOX patterning in vivo, in vitro approaches for differentiating human pluripotent stem cells (hPSCs) to posterior neural fates coarsely pattern HOX expression thereby generating cultures broadly specified to hindbrain or spinal cord regions. Here, we demonstrate that successive activation of fibroblast growth factor, Wnt/β-catenin, and growth differentiation factor signaling during hPSC differentiation generates stable, homogenous SOX2+/Brachyury+ neuromesoderm that exhibits progressive, full colinear HOX activation over 7 days. Switching to retinoic acid treatment at any point during this process halts colinear HOX activation and transitions the neuromesoderm into SOX2+/PAX6+ neuroectoderm with predictable, discrete HOX gene/protein profiles that can be further differentiated into region-specific cells, e.g., motor neurons. This fully defined approach significantly expands capabilities to derive regional neural phenotypes from diverse hindbrain and spinal cord domains. Deterministic HOX expression in hPSC-derived neuromesoderm progenitors (NMPs) Wnt/β-catenin, FGF, and GDF signaling regulate HOX activation in NMPs Retinoic acid (RA) transitions NMPs to neuroectoderm and halts HOX activation Neural cells can be patterned to any rostrocaudal hindbrain or spinal cord domain
Collapse
|
49
|
Lovrics A, Gao Y, Juhász B, Bock I, Byrne HM, Dinnyés A, Kovács KA. Boolean modelling reveals new regulatory connections between transcription factors orchestrating the development of the ventral spinal cord. PLoS One 2014; 9:e111430. [PMID: 25398016 PMCID: PMC4232242 DOI: 10.1371/journal.pone.0111430] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 08/26/2014] [Indexed: 11/19/2022] Open
Abstract
We have assembled a network of cell-fate determining transcription factors that play a key role in the specification of the ventral neuronal subtypes of the spinal cord on the basis of published transcriptional interactions. Asynchronous Boolean modelling of the network was used to compare simulation results with reported experimental observations. Such comparison highlighted the need to include additional regulatory connections in order to obtain the fixed point attractors of the model associated with the five known progenitor cell types located in the ventral spinal cord. The revised gene regulatory network reproduced previously observed cell state switches between progenitor cells observed in knock-out animal models or in experiments where the transcription factors were overexpressed. Furthermore the network predicted the inhibition of Irx3 by Nkx2.2 and this prediction was tested experimentally. Our results provide evidence for the existence of an as yet undescribed inhibitory connection which could potentially have significance beyond the ventral spinal cord. The work presented in this paper demonstrates the strength of Boolean modelling for identifying gene regulatory networks.
Collapse
Affiliation(s)
| | - Yu Gao
- Biotalentum Ltd., Gödöllö, Hungary
| | | | - István Bock
- Biotalentum Ltd., Gödöllö, Hungary
- Molecular Animal Biotechnology Laboratory, Szent Istvan University, Gödöllö, Hungary
| | - Helen M. Byrne
- Oxford Centre for Collaborative Applied Mathematics, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - András Dinnyés
- Biotalentum Ltd., Gödöllö, Hungary
- Molecular Animal Biotechnology Laboratory, Szent Istvan University, Gödöllö, Hungary
- Department of Farm Animal Health, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Krisztián A. Kovács
- Biotalentum Ltd., Gödöllö, Hungary
- Institute of Science and Technology, Klosterneuburg, Austria
| |
Collapse
|
50
|
Quiroga AC, Stolt CC, Diez del Corral R, Dimitrov S, Pérez-Alcalá S, Sock E, Barbas JA, Wegner M, Morales AV. Sox5 controls dorsal progenitor and interneuron specification in the spinal cord. Dev Neurobiol 2014; 75:522-38. [PMID: 25363628 DOI: 10.1002/dneu.22240] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 10/22/2014] [Accepted: 10/25/2014] [Indexed: 11/08/2022]
Abstract
The basic organization of somatosensory circuits in the spinal cord is already setup during the initial patterning of the dorsal neural tube. Extrinsic signals, such as Wnt and TGF-β pathways, activate combinatorial codes of transcription factors that are responsible for generating a pattern of discrete domains of dorsal progenitors (dp). These progenitors will give rise to distinct dorsal interneurons (dI). The Wnt/ βcatenin signaling pathway controls specification of dp/dI1-3 progenitors and interneurons. According to the current model in the field, Wnt/βcatenin activity seems to act in a graded fashion in the spinal cord, as different relative levels determine the identity of adjacent progenitors. However, it is not clear how this activity gradient is controlled and how the identities of dI1-3 are differentially regulated by Wnt signalling. We have determined that two SoxD transcription factors, Sox5 and Sox6, are expressed in restricted domains of dorsal progenitors in the neural tube. Using gain- and loss-of function approaches in chicken embryos, we have established that Sox5 controls cell fate specification of dp2 and dp3 progenitors and, as a result, controls the correct number of the corresponding dorsal interneurons (dI2 and dI3). Furthermore, Sox5 exerts its function by restricting dorsally Wnt signaling activity via direct transcriptional induction of the negative Wnt pathway regulator Axin2. By that way, Sox5 acts as a Wnt pathway modulator that contributes to sharpen the dorsal gradient of Wnt/βcatenin activity to control the distinction of two functionally distinct types of interneurons, dI2 and dI3 involved in the somatosensory relay.
Collapse
Affiliation(s)
- Alejandra C Quiroga
- Department of Cellular, Molecular and Developmental Neurobiology, Instituto Cajal, CSIC, Madrid, 28002, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|