1
|
Blair JD, Hartman A, Zenk F, Wahle P, Brancati G, Dalgarno C, Treutlein B, Satija R. Phospho-seq: integrated, multi-modal profiling of intracellular protein dynamics in single cells. Nat Commun 2025; 16:1346. [PMID: 39905064 DOI: 10.1038/s41467-025-56590-7] [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: 11/15/2024] [Accepted: 01/22/2025] [Indexed: 02/06/2025] Open
Abstract
Cell signaling plays a critical role in neurodevelopment, regulating cellular behavior and fate. While multimodal single-cell sequencing technologies are rapidly advancing, scalable and flexible profiling of cell signaling states alongside other molecular modalities remains challenging. Here we present Phospho-seq, an integrated approach that aims to quantify cytoplasmic and nuclear proteins, including those with post-translational modifications, and to connect their activity with cis-regulatory elements and transcriptional targets. We utilize a simplified benchtop antibody conjugation method to create large custom neuro-focused antibody panels for simultaneous protein and scATAC-seq profiling on whole cells, alongside both experimental and computational strategies to incorporate transcriptomic measurements. We apply our workflow to cell lines, induced pluripotent stem cells, and months-old retinal and brain organoids to demonstrate its broad applicability. We show that Phospho-seq can provide insights into cellular states and trajectories, shed light on gene regulatory relationships, and help explore the causes and effects of diverse cell signaling in neurodevelopment.
Collapse
Affiliation(s)
- John D Blair
- New York Genome Center, New York, NY, USA
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | | | | | | | | | | | | | - Rahul Satija
- New York Genome Center, New York, NY, USA.
- Center for Genomics and Systems Biology, New York University, New York, NY, USA.
| |
Collapse
|
2
|
Schubert FR, Dietrich S. Naturally occurring, rostrally conjoining chicken twins attempt to make a forebrain. Dev Biol 2025; 520:171-179. [PMID: 39848482 DOI: 10.1016/j.ydbio.2025.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 01/07/2025] [Accepted: 01/18/2025] [Indexed: 01/25/2025]
Abstract
Conjoined twinning is a special case of monozygotic, monoamniotic twinning. Human conjoined twinning, and vertebrate conjoined twinning in general, is a very rare phenomenon. It has been suggested that the risk of conjoined twinning increases with some medication and upon assisted reproduction. Survival rates are low. When conjoined twins occur in the chicken, they most often present with fused heads, anatomically unrecognisable brains and two normal bodies. Recent studies suggested that forebrain, midbrain and rostral hindbrain identities are established in the early epiblast before neural induction and independent from caudal hindbrain and spinal cord identities. Therefore, it is unclear whether in conjoined twins, the aberrant brain anatomy is a result of the rostral fusion, or whether the brains failed to develop in the first place. Here, we collected conjoined twins as they spontaneously appeared in eggs incubated for stages HH4 (late primitive steak stage) to HH13 (early pharyngula). The twinned embryos and stage-matched normal embryos were analysed for the expression of the rostral epiblast and forebrain-midbrain marker Otx2 and the ventral forebrain marker Six3. We found normal anatomy and marker gene expression that lasted up to stage HH9. By HH12-13, the brain anatomy had deteriorated, but marker genes remained expressed. This suggests that the fusing embryos attempted to generate a brain including the forebrain. Besides addressing forebrain development, our work for the first time provides a time frame to study the mechanisms underlying the interaction and fusion of conjoined twins, which will pave the way to a better understanding and management of risk factors in humans.
Collapse
Affiliation(s)
- Frank R Schubert
- Institute of Life Sciences and Health (ILSH), School of the Environment and Life Sciences (SELS), University of Portsmouth, Portsmouth, PO1 2DY, UK
| | - Susanne Dietrich
- Institute of Life Sciences and Health (ILSH), School of Medicine, Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, PO1 2DT, UK.
| |
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
|
Elder NH, Majd A, Bulger EA, Samuel RM, Zholudeva LV, McDevitt TC, Fattahi F. Distinct differentiation trajectories leave lasting impacts on gene regulation and function of V2a interneurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.03.626573. [PMID: 39677634 PMCID: PMC11642877 DOI: 10.1101/2024.12.03.626573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
During development, early regionalization segregates lineages and directs diverse cell fates. Sometimes, however, distinct progenitors produce analogous cell types. For example, V2a neurons, are excitatory interneurons that emerge from different anteroposterior progenitors. V2a neurons demonstrate remarkable plasticity after spinal cord injury and improve motor function, showing potential for cell therapy. To examine how lineage origins shape their properties, we differentiated V2a neurons from hPSC-derived progenitors with distinct anteroposterior identities. Single-nucleus multiomic analysis revealed lineage-specific transcription factor motifs and numerous differentially expressed genes related to axon growth and calcium handling. Bypassing lineage patterning via transcription factor-induced differentiation yielded neurons distinct from both developmentally relevant populations and human tissue, emphasizing the need to follow developmental steps to generate authentic cell identities. Using in silico and in vitro loss-of-function analyses, we identified CREB5 and TCF7L2 as regulators specific to posterior identities, underscoring the critical role of lieage origins in determining cell states and functions.
Collapse
Affiliation(s)
- Nicholas H. Elder
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94143, USA
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, CA 94158, USA
| | - Alireza Majd
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Emily A. Bulger
- Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, CA 94158, USA
- Gladstone Institute of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA 94158, USA
- Current address: Genentech, South San Francisco, California 94080 USA
| | - Ryan M. Samuel
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Lyandysha V. Zholudeva
- Gladstone Institute of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Todd C. McDevitt
- Gladstone Institute of Cardiovascular Disease, Gladstone Institutes, San Francisco, CA 94158, USA
- Current address: Genentech, South San Francisco, California 94080 USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Faranak Fattahi
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, 94143, USA
- Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA 94110, USA
| |
Collapse
|
5
|
Karkali K, Martín-Blanco E. The evolutionary and mechanical principles shaping the Drosophila embryonic ventral nerve cord. Cells Dev 2024; 180:203973. [PMID: 39490740 DOI: 10.1016/j.cdev.2024.203973] [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: 05/22/2024] [Revised: 09/19/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
The establishment of communication circuits requires bringing sources and targets into contact, either directly or indirectly. The Central Nervous System (CNS)'s ability to interpret the environment and generate precise responses depends on the functional efficiency of its neural network, which in turn relies on the 3D spatial organization of its constituents, mainly neurons and glia. Throughout evolution, sensory integration and motor response coordination became linked with the merging of the brain and nerve cord (NC) in the urbilaterian CNS. In most arthropods, the NC follows a specific topological plan and consists of a fixed number of neuromeres (thoracic and abdominal ganglia with commissural interconnections and a single terminal ganglion). The number, spacing, and fusion of neuromeres are species-specific and can change during embryogenesis or post-embryonic life. During Drosophila embryogenesis, the NC condenses along the Anterior-Posterior (AP) axis in a stereotypical manner, bringing neuromeres closer together. This process has revealed several key parameters, including its morphogenetic mechanics, the roles of various cellular, molecular, and structural components, and the functional purpose of its balanced design. The embryonic NC serves as a valuable model for investigating the ancient mechanisms underlying the structural organization, sensory integration, and motor coordination of the CNS. While many aspects of ganglionic fusion remain unknown, ongoing research promises to provide a more comprehensive understanding of the mechanical and evolutionary principles that govern it.
Collapse
Affiliation(s)
- Katerina Karkali
- Instituto de Biología Molecular de Barcelona (CSIC), Parc Cientific de Barcelona, Baldiri Reixac 10-12, 08028 Barcelona, Spain.
| | - Enrique Martín-Blanco
- Instituto de Biología Molecular de Barcelona (CSIC), Parc Cientific de Barcelona, Baldiri Reixac 10-12, 08028 Barcelona, Spain
| |
Collapse
|
6
|
Grass T, Dokuzluoglu Z, Rodríguez-Muela N. Neuromuscular Organoids to Study Spinal Cord Development and Disease. Methods Mol Biol 2024. [PMID: 39570548 DOI: 10.1007/7651_2024_574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
Many aspects of neurodegenerative disease pathology remain unresolved. Why do certain neuronal subpopulations acquire vulnerability to stress or mutations in ubiquitously expressed genes, while others remain resilient? Do these neurons harbor intrinsic marks that make them prone to degeneration? Do these diseases have a neurodevelopmental component? Lacking this fundamental knowledge hampers the discovery of efficacious treatments. While it is well established that human organoids enable the modeling of brain-related diseases, we still lack an organoid model that recapitulates the regionalization complexity and physiology of the spinal cord. Here, we describe an advanced experimental protocol to generate neuromuscular organoids composed of a wide rostro-caudal (RC) diversity of spinal motor neurons (spMNs) and mesodermal progenitor-derived muscle cells. This model therefore allows for the robust and reproducible study of neuromuscular unit development and disease.
Collapse
Affiliation(s)
- Tobias Grass
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany
| | - Zeynep Dokuzluoglu
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany
| | - Natalia Rodríguez-Muela
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany.
- Technische Universität Dresden (TUD), Center for Regenerative Therapies Dresden, Dresden, Germany.
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany.
| |
Collapse
|
7
|
Chen S, Hayoun-Neeman D, Nagar M, Pinyan S, Hadad L, Yaacobov L, Alon L, Shachar LE, Swissa T, Kryukov O, Gershoni-Yahalom O, Rosental B, Cohen S, Lichtenstein RG. Terminal α1,2-fucosylation of glycosphingolipids by FUT1 is a key regulator in early cell-fate decisions. EMBO Rep 2024; 25:4433-4464. [PMID: 39256596 PMCID: PMC11467398 DOI: 10.1038/s44319-024-00243-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 08/09/2024] [Accepted: 08/23/2024] [Indexed: 09/12/2024] Open
Abstract
The embryonic cell surface is rich in glycosphingolipids (GSLs), which change during differentiation. The reasons for GSL subgroup variation during early embryogenesis remain elusive. By combining genomic approaches, flow cytometry, confocal imaging, and transcriptomic data analysis, we discovered that α1,2-fucosylated GSLs control the differentiation of human pluripotent cells (hPCs) into germ layer tissues. Overexpression of α1,2-fucosylated GSLs disrupts hPC differentiation into mesodermal lineage and reduces differentiation into cardiomyocytes. Conversely, reducing α1,2-fucosylated groups promotes hPC differentiation and mesoderm commitment in response to external signals. We find that bone morphogenetic protein 4 (BMP4), a mesodermal gene inducer, suppresses α1,2-fucosylated GSL expression. Overexpression of α1,2-fucosylated GSLs impairs SMAD activation despite BMP4 presence, suggesting α-fucosyl end groups as BMP pathway regulators. Additionally, the absence of α1,2-fucosylated GSLs in early/late mesoderm and primitive streak stages in mouse embryos aligns with the hPC results. Thus, α1,2-fucosylated GSLs may regulate early cell-fate decisions and embryo development by modulating cell signaling.
Collapse
Affiliation(s)
- Saray Chen
- Avram and Stella Goren-Goldstein Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Dana Hayoun-Neeman
- Avram and Stella Goren-Goldstein Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Michal Nagar
- Avram and Stella Goren-Goldstein Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Sapir Pinyan
- Avram and Stella Goren-Goldstein Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Limor Hadad
- Avram and Stella Goren-Goldstein Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Liat Yaacobov
- Avram and Stella Goren-Goldstein Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Lilach Alon
- Avram and Stella Goren-Goldstein Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Liraz Efrat Shachar
- Avram and Stella Goren-Goldstein Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Tair Swissa
- Avram and Stella Goren-Goldstein Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Olga Kryukov
- Avram and Stella Goren-Goldstein Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Orly Gershoni-Yahalom
- Regenerative Medicine and Stem Cell (RMSC) Research Center, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Benyamin Rosental
- Regenerative Medicine and Stem Cell (RMSC) Research Center, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Smadar Cohen
- Avram and Stella Goren-Goldstein Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- Regenerative Medicine and Stem Cell (RMSC) Research Center, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
- The Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | - Rachel G Lichtenstein
- Avram and Stella Goren-Goldstein Department of Biotechnology Engineering, Faculty of Engineering Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel.
- Regenerative Medicine and Stem Cell (RMSC) Research Center, Ben-Gurion University of the Negev, Beer-Sheva, 8410501, Israel.
| |
Collapse
|
8
|
El Azhar Y, Schulthess P, van Oostrom MJ, Weterings SDC, Meijer WHM, Tsuchida-Straeten N, Thomas WM, Bauer M, Sonnen KF. Unravelling differential Hes1 dynamics during axis elongation of mouse embryos through single-cell tracking. Development 2024; 151:dev202936. [PMID: 39315665 DOI: 10.1242/dev.202936] [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: 04/08/2024] [Accepted: 08/22/2024] [Indexed: 09/25/2024]
Abstract
The intricate dynamics of Hes expression across diverse cell types in the developing vertebrate embryonic tail have remained elusive. To address this, we have developed an endogenously tagged Hes1-Achilles mouse line, enabling precise quantification of dynamics at the single-cell resolution across various tissues. Our findings reveal striking disparities in Hes1 dynamics between presomitic mesoderm (PSM) and preneural tube (pre-NT) cells. While pre-NT cells display variable, low-amplitude oscillations, PSM cells exhibit synchronized, high-amplitude oscillations. Upon the induction of differentiation, the oscillation amplitude increases in pre-NT cells. Additionally, our study of Notch inhibition on Hes1 oscillations unveils distinct responses in PSM and pre-NT cells, corresponding to differential Notch ligand expression dynamics. These findings suggest the involvement of separate mechanisms driving Hes1 oscillations. Thus, Hes1 demonstrates dynamic behaviour across adjacent tissues of the embryonic tail, yet the varying oscillation parameters imply differences in the information that can be transmitted by these dynamics.
Collapse
Affiliation(s)
- Yasmine El Azhar
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), University Medical Center Utrecht, Utrecht 3584, The Netherlands
| | - Pascal Schulthess
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), University Medical Center Utrecht, Utrecht 3584, The Netherlands
| | - Marek J van Oostrom
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), University Medical Center Utrecht, Utrecht 3584, The Netherlands
| | - Sonja D C Weterings
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), University Medical Center Utrecht, Utrecht 3584, The Netherlands
| | - Wilke H M Meijer
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), University Medical Center Utrecht, Utrecht 3584, The Netherlands
| | | | - Wouter M Thomas
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), University Medical Center Utrecht, Utrecht 3584, The Netherlands
| | - Marianne Bauer
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), University Medical Center Utrecht, Utrecht 3584, The Netherlands
| | - Katharina F Sonnen
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), University Medical Center Utrecht, Utrecht 3584, The Netherlands
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Technical University of Delft, Van der Maasweg 9, 2629 HZ Delft, The Netherlands
| |
Collapse
|
9
|
Grass T, Dokuzluoglu Z, Buchner F, Rosignol I, Thomas J, Caldarelli A, Dalinskaya A, Becker J, Rost F, Marass M, Wirth B, Beyer M, Bonaguro L, Rodriguez-Muela N. Isogenic patient-derived organoids reveal early neurodevelopmental defects in spinal muscular atrophy initiation. Cell Rep Med 2024; 5:101659. [PMID: 39067446 PMCID: PMC11384962 DOI: 10.1016/j.xcrm.2024.101659] [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: 07/25/2023] [Revised: 02/26/2024] [Accepted: 07/02/2024] [Indexed: 07/30/2024]
Abstract
Whether neurodevelopmental defects underlie postnatal neuronal death in neurodegeneration is an intriguing hypothesis only recently explored. Here, we focus on spinal muscular atrophy (SMA), a neuromuscular disorder caused by reduced survival of motor neuron (SMN) protein levels leading to spinal motor neuron (MN) loss and muscle wasting. Using the first isogenic patient-derived induced pluripotent stem cell (iPSC) model and a spinal cord organoid (SCO) system, we show that SMA SCOs exhibit abnormal morphological development, reduced expression of early neural progenitor markers, and accelerated expression of MN progenitor and MN markers. Longitudinal single-cell RNA sequencing reveals marked defects in neural stem cell specification and fewer MNs, favoring mesodermal progenitors and muscle cells, a bias also seen in early SMA mouse embryos. Surprisingly, SMN2-to-SMN1 conversion does not fully reverse these developmental abnormalities. These suggest that early neurodevelopmental defects may underlie later MN degeneration, indicating that postnatal SMN-increasing interventions might not completely amend SMA pathology in all patients.
Collapse
Affiliation(s)
- Tobias Grass
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany.
| | - Zeynep Dokuzluoglu
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany
| | - Felix Buchner
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany
| | - Ines Rosignol
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany; Technische Universität Dresden (TUD), Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Joshua Thomas
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany
| | - Antonio Caldarelli
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany
| | - Anna Dalinskaya
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany
| | - Jutta Becker
- Institute of Human Genetics, University Hospital of Cologne, Cologne, Germany
| | - Fabian Rost
- DRESDEN-concept Genome Center, Technology Platform at the Center for Molecular and Cellular Bioengineering, TUD, Dresden, Germany
| | - Michele Marass
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany; Center for Systems Biology Dresden, Dresden, Germany
| | - Brunhilde Wirth
- Institute of Human Genetics, University Hospital of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Center for Rare Diseases, University Hospital of Cologne, Cologne, Germany
| | - Marc Beyer
- Systems Medicine, DZNE, Bonn, Germany; PRECISE Platform for Single Cell Genomics and Epigenomics, DZNE & University of Bonn and West German Genome Center, Bonn, Germany; Immunogenomics & Neurodegeneration, DZNE, Bonn, Germany
| | - Lorenzo Bonaguro
- Systems Medicine, DZNE, Bonn, Germany; Genomics & Immunoregulation, LIMES Institute, University of Bonn, Bonn, Germany
| | - Natalia Rodriguez-Muela
- German Center for Neurodegenerative Diseases e.V. (DZNE), Dresden, Germany; Technische Universität Dresden (TUD), Center for Regenerative Therapies Dresden, Dresden, Germany; Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany.
| |
Collapse
|
10
|
Xue X, Kim YS, Ponce-Arias AI, O'Laughlin R, Yan RZ, Kobayashi N, Tshuva RY, Tsai YH, Sun S, Zheng Y, Liu Y, Wong FCK, Surani A, Spence JR, Song H, Ming GL, Reiner O, Fu J. A patterned human neural tube model using microfluidic gradients. Nature 2024; 628:391-399. [PMID: 38408487 PMCID: PMC11006583 DOI: 10.1038/s41586-024-07204-7] [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: 01/26/2022] [Accepted: 02/16/2024] [Indexed: 02/28/2024]
Abstract
The human nervous system is a highly complex but organized organ. The foundation of its complexity and organization is laid down during regional patterning of the neural tube, the embryonic precursor to the human nervous system. Historically, studies of neural tube patterning have relied on animal models to uncover underlying principles. Recently, models of neurodevelopment based on human pluripotent stem cells, including neural organoids1-5 and bioengineered neural tube development models6-10, have emerged. However, such models fail to recapitulate neural patterning along both rostral-caudal and dorsal-ventral axes in a three-dimensional tubular geometry, a hallmark of neural tube development. Here we report a human pluripotent stem cell-based, microfluidic neural tube-like structure, the development of which recapitulates several crucial aspects of neural patterning in brain and spinal cord regions and along rostral-caudal and dorsal-ventral axes. This structure was utilized for studying neuronal lineage development, which revealed pre-patterning of axial identities of neural crest progenitors and functional roles of neuromesodermal progenitors and the caudal gene CDX2 in spinal cord and trunk neural crest development. We further developed dorsal-ventral patterned microfluidic forebrain-like structures with spatially segregated dorsal and ventral regions and layered apicobasal cellular organizations that mimic development of the human forebrain pallium and subpallium, respectively. Together, these microfluidics-based neurodevelopment models provide three-dimensional lumenal tissue architectures with in vivo-like spatiotemporal cell differentiation and organization, which will facilitate the study of human neurodevelopment and disease.
Collapse
Affiliation(s)
- Xufeng Xue
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Yung Su Kim
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Alfredo-Isaac Ponce-Arias
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Richard O'Laughlin
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Robin Zhexuan Yan
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Norio Kobayashi
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Rami Yair Tshuva
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Yu-Hwai Tsai
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Shiyu Sun
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Yi Zheng
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Yue Liu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Frederick C K Wong
- Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
| | - Azim Surani
- Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Jason R Spence
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, USA
- Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
- The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Orly Reiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA.
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
11
|
Ortiz-Salazar MA, Camacho-Aguilar E, Warmflash A. Endogenous Nodal switches Wnt interpretation from posteriorization to germ layer differentiation in geometrically constrained human pluripotent cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.13.584912. [PMID: 38559061 PMCID: PMC10979992 DOI: 10.1101/2024.03.13.584912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The Wnt pathway is essential for inducing the primitive streak, the precursor of the mesendoderm, as well as setting anterior-posterior coordinates. How Wnt coordinates these diverse activities remains incompletely understood. Here, we show that in Wnt-treated human pluripotent cells, endogenous Nodal signaling is a crucial switch between posteriorizing and primitive streak-including activities. While treatment with Wnt posteriorizes cells in standard culture, in micropatterned colonies, higher levels of endogenously induced Nodal signaling combine with exogenous Wnt to drive endoderm differentiation. Inhibition of Nodal signaling restores dose-dependent posteriorization by Wnt. In the absence of Nodal inhibition, micropatterned colonies undergo spontaneous, elaborate morphogenesis concomitant with endoderm differentiation even in the absence of added extracellular matrix proteins like Matrigel. Our study shows how Wnt and Nodal combinatorially coordinate germ layer differentiation with AP patterning and establishes a system to study a natural self-organizing morphogenetic event in in vitro culture.
Collapse
Affiliation(s)
| | - Elena Camacho-Aguilar
- Department of Biosciences, Rice University, Houston, TX, USA 77005
- Present address: Department of Gene Regulation and Morphogenesis, Andalusian Center for Developmental Biology (CSIC-UPO-JA), Seville, Spain, 41013
| | - Aryeh Warmflash
- Department of Biosciences, Rice University, Houston, TX, USA 77005
- Department of Bioengineering, Rice University, Houston, TX, USA 77005
| |
Collapse
|
12
|
Pappas MP, Kawakami H, Corcoran D, Chen KQ, Scott EP, Wong J, Gearhart MD, Nishinakamura R, Nakagawa Y, Kawakami Y. Sall4 regulates posterior trunk mesoderm development by promoting mesodermal gene expression and repressing neural genes in the mesoderm. Development 2024; 151:dev202649. [PMID: 38345319 PMCID: PMC10946440 DOI: 10.1242/dev.202649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 01/30/2024] [Indexed: 03/05/2024]
Abstract
The trunk axial skeleton develops from paraxial mesoderm cells. Our recent study demonstrated that conditional knockout of the stem cell factor Sall4 in mice by TCre caused tail truncation and a disorganized axial skeleton posterior to the lumbar level. Based on this phenotype, we hypothesized that, in addition to the previously reported role of Sall4 in neuromesodermal progenitors, Sall4 is involved in the development of the paraxial mesoderm tissue. Analysis of gene expression and SALL4 binding suggests that Sall4 directly or indirectly regulates genes involved in presomitic mesoderm differentiation, somite formation and somite differentiation. Furthermore, ATAC-seq in TCre; Sall4 mutant posterior trunk mesoderm shows that Sall4 knockout reduces chromatin accessibility. We found that Sall4-dependent open chromatin status drives activation and repression of WNT signaling activators and repressors, respectively, to promote WNT signaling. Moreover, footprinting analysis of ATAC-seq data suggests that Sall4-dependent chromatin accessibility facilitates CTCF binding, which contributes to the repression of neural genes within the mesoderm. This study unveils multiple mechanisms by which Sall4 regulates paraxial mesoderm development by directing activation of mesodermal genes and repression of neural genes.
Collapse
Affiliation(s)
- Matthew P. Pappas
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Hiroko Kawakami
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
- Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Dylan Corcoran
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Katherine Q. Chen
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Earl Parker Scott
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Julia Wong
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Micah D. Gearhart
- Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Obstetrics, Gynecology and Women's Health, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Yasushi Nakagawa
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
- Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Yasuhiko Kawakami
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
- Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
13
|
Yang A, Chidiac R, Russo E, Steenland H, Pauli Q, Bonin R, Blazer LL, Adams JJ, Sidhu SS, Goeva A, Salahpour A, Angers S. Exploiting spatiotemporal regulation of FZD5 during neural patterning for efficient ventral midbrain specification. Development 2024; 151:dev202545. [PMID: 38358799 PMCID: PMC10946437 DOI: 10.1242/dev.202545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 02/06/2024] [Indexed: 02/16/2024]
Abstract
The Wnt/β-catenin signaling governs anterior-posterior neural patterning during development. Current human pluripotent stem cell (hPSC) differentiation protocols use a GSK3 inhibitor to activate Wnt signaling to promote posterior neural fate specification. However, GSK3 is a pleiotropic kinase involved in multiple signaling pathways and, as GSK3 inhibition occurs downstream in the signaling cascade, it bypasses potential opportunities for achieving specificity or regulation at the receptor level. Additionally, the specific roles of individual FZD receptors in anterior-posterior patterning are poorly understood. Here, we have characterized the cell surface expression of FZD receptors in neural progenitor cells with different regional identity. Our data reveal unique upregulation of FZD5 expression in anterior neural progenitors, and this expression is downregulated as cells adopt a posterior fate. This spatial regulation of FZD expression constitutes a previously unreported regulatory mechanism that adjusts the levels of β-catenin signaling along the anterior-posterior axis and possibly contributes to midbrain-hindbrain boundary formation. Stimulation of Wnt/β-catenin signaling in hPSCs, using a tetravalent antibody that selectively triggers FZD5 and LRP6 clustering, leads to midbrain progenitor differentiation and gives rise to functional dopaminergic neurons in vitro and in vivo.
Collapse
Affiliation(s)
- Andy Yang
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Rony Chidiac
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Emma Russo
- Department of Pharmacology and Toxicology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Hendrik Steenland
- Department of Pharmacology and Toxicology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- NeuroTek Innovative Technology, Toronto, ON M6C 3A2, Canada
| | - Quinn Pauli
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Robert Bonin
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Levi L. Blazer
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Jarrett J. Adams
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
| | - Sachdev S. Sidhu
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Aleksandrina Goeva
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Ali Salahpour
- Department of Pharmacology and Toxicology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Stephane Angers
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1, Canada
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
- Department of Biochemistry, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
14
|
Abstract
In avian and mammalian embryos the "organizer" property associated with neural induction of competent ectoderm into a neural plate and its subsequent patterning into rostro-caudal domains resides at the tip of the primitive streak before neurulation begins, and before a morphological Hensen's node is discernible. The same region and its later derivatives (like the notochord) also have the ability to "dorsalize" the adjacent mesoderm, for example by converting lateral plate mesoderm into paraxial (pre-somitic) mesoderm. Both neural induction and dorsalization of the mesoderm involve inhibition of BMP, and the former also requires other signals. This review surveys the key experiments done to elucidate the functions of the organizer and the mechanisms of neural induction in amniotes. We conclude that the mechanisms of neural induction in amniotes and anamniotes are likely to be largely the same; apparent differences are likely to be due to differences in experimental approaches dictated by embryo topology and other practical constraints. We also discuss the relationships between "neural induction" assessed by grafts of the organizer and normal neural plate development, as well as how neural induction relates to the generation of neuronal cells from embryonic and other stem cells in vitro.
Collapse
Affiliation(s)
- Claudio D Stern
- Department of Cell and Developmental Biology, University College London, London, United Kingdom.
| |
Collapse
|
15
|
Nakamura K, Watanabe Y, Boitet C, Satake S, Iida H, Yoshihi K, Ishii Y, Kato K, Kondoh H. Wnt signal-dependent antero-posterior specification of early-stage CNS primordia modeled in EpiSC-derived neural stem cells. Front Cell Dev Biol 2024; 11:1260528. [PMID: 38405136 PMCID: PMC10884098 DOI: 10.3389/fcell.2023.1260528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 11/30/2023] [Indexed: 02/27/2024] Open
Abstract
The specification of the embryonic central nervous system (CNS) into future brain (forebrain, midbrain, or hindbrain) and spinal cord (SC) regions is a critical step of CNS development. A previous chicken embryo study indicated that anterior epiblast cells marked by Sox2 N2 enhancer activity are specified to the respective brain regions during the transition phase of the epiblast to the neural plate-forming neural primordium. The present study showed that the SC precursors positioned posterior to the hindbrain precursors in the anterior epiblast migrated posteriorly in contrast to the anterior migration of brain precursors. The anteroposterior specification of the CNS precursors occurs at an analogous time (∼E7.5) in mouse embryos, in which an anterior-to-posterior incremental gradient of Wnt signal strength was observed. To examine the possible Wnt signal contribution to the anteroposterior CNS primordium specification, we utilized mouse epiblast stem cell (EpiSC)-derived neurogenesis in culture. EpiSCs maintained in an activin- and FGF2-containing medium start neural development after the removal of activin, following a day in a transitory state. We placed activin-free EpiSCs in EGF- and FGF2-containing medium to arrest neural development and expand the cells into neural stem cells (NSCs). Simultaneously, a Wnt antagonist or agonist was added to the culture, with the anticipation that different levels of Wnt signals would act on the transitory cells to specify CNS regionality; then, the Wnt-treated cells were expanded as NSCs. Gene expression profiles of six NSC lines were analyzed using microarrays and single-cell RNA-seq. The NSC lines demonstrated anteroposterior regional specification in response to increasing Wnt signal input levels: forebrain-midbrain-, hindbrain-, cervical SC-, and thoracic SC-like lines. The regional coverage of these NSC lines had a range; for instance, the XN1 line expressed Otx2 and En2, indicating midbrain characteristics, but additionally expressed the SC-characteristic Hoxa5. The ranges in the anteroposterior specification of neural primordia may be narrowed as neural development proceeds. The thoracic SC is presumably the posterior limit of the contribution by anterior epiblast-derived neural progenitors, as the characteristics of more posterior SC regions were not displayed.
Collapse
Affiliation(s)
- Kae Nakamura
- Faculty of Life Sciences, Kyoto Sangyo University, Kita-ku, Kyoto, Japan
| | - Yusaku Watanabe
- Faculty of Life Sciences, Kyoto Sangyo University, Kita-ku, Kyoto, Japan
| | - Claire Boitet
- Faculty of Life Sciences, Kyoto Sangyo University, Kita-ku, Kyoto, Japan
- Université Joseph Fourier, Domaine Universitaire, Saint-Martin-d’Hères, France
| | - Sayaka Satake
- Faculty of Life Sciences, Kyoto Sangyo University, Kita-ku, Kyoto, Japan
| | - Hideaki Iida
- Faculty of Life Sciences, Kyoto Sangyo University, Kita-ku, Kyoto, Japan
| | - Koya Yoshihi
- Faculty of Life Sciences, Kyoto Sangyo University, Kita-ku, Kyoto, Japan
| | - Yasuo Ishii
- Faculty of Life Sciences, Kyoto Sangyo University, Kita-ku, Kyoto, Japan
- Department of Biology, School of Medicine, Tokyo Women’s Medical University, Tokyo, Japan
| | - Kagayaki Kato
- National Institute for Basic Biology, Okazaki, Aichi, Japan
| | - Hisato Kondoh
- Faculty of Life Sciences, Kyoto Sangyo University, Kita-ku, Kyoto, Japan
- Biohistory Research Hall, Takatsuki, Osaka, Japan
| |
Collapse
|
16
|
Cooper F, Souilhol C, Haston S, Gray S, Boswell K, Gogolou A, Frith TJR, Stavish D, James BM, Bose D, Kim Dale J, Tsakiridis A. Notch signalling influences cell fate decisions and HOX gene induction in axial progenitors. Development 2024; 151:dev202098. [PMID: 38223992 PMCID: PMC10911136 DOI: 10.1242/dev.202098] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 12/20/2023] [Indexed: 01/16/2024]
Abstract
The generation of the post-cranial embryonic body relies on the coordinated production of spinal cord neurectoderm and presomitic mesoderm cells from neuromesodermal progenitors (NMPs). This process is orchestrated by pro-neural and pro-mesodermal transcription factors that are co-expressed in NMPs together with Hox genes, which are essential for axial allocation of NMP derivatives. NMPs reside in a posterior growth region, which is marked by the expression of Wnt, FGF and Notch signalling components. Although the importance of Wnt and FGF in influencing the induction and differentiation of NMPs is well established, the precise role of Notch remains unclear. Here, we show that the Wnt/FGF-driven induction of NMPs from human embryonic stem cells (hESCs) relies on Notch signalling. Using hESC-derived NMPs and chick embryo grafting, we demonstrate that Notch directs a pro-mesodermal character at the expense of neural fate. We show that Notch also contributes to activation of HOX gene expression in human NMPs, partly in a non-cell-autonomous manner. Finally, we provide evidence that Notch exerts its effects via the establishment of a negative-feedback loop with FGF signalling.
Collapse
Affiliation(s)
- Fay Cooper
- School of Biosciences, The University of Sheffield, Sheffield S10 2TN, UK
- Neuroscience Institute, The University of Sheffield, Sheffield S10 2TN, UK
| | - Celine Souilhol
- School of Biosciences, The University of Sheffield, Sheffield S10 2TN, UK
- Neuroscience Institute, The University of Sheffield, Sheffield S10 2TN, UK
- Biomolecular Sciences Research Centre, Department of Biosciences and Chemistry, Sheffield Hallam University, Sheffield S1 1WB, UK
| | - Scott Haston
- Developmental Biology and Cancer, Birth Defects Research Centre, UCL GOS Institute of Child Health, London WC1N 1EH, UK
- Division of Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee DD1 4HN, UK
| | - Shona Gray
- Division of Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee DD1 4HN, UK
| | - Katy Boswell
- School of Biosciences, The University of Sheffield, Sheffield S10 2TN, UK
- Neuroscience Institute, The University of Sheffield, Sheffield S10 2TN, UK
| | - Antigoni Gogolou
- School of Biosciences, The University of Sheffield, Sheffield S10 2TN, UK
- Neuroscience Institute, The University of Sheffield, Sheffield S10 2TN, UK
| | - Thomas J. R. Frith
- School of Biosciences, The University of Sheffield, Sheffield S10 2TN, UK
- Neuroscience Institute, The University of Sheffield, Sheffield S10 2TN, UK
| | - Dylan Stavish
- School of Biosciences, The University of Sheffield, Sheffield S10 2TN, UK
- Neuroscience Institute, The University of Sheffield, Sheffield S10 2TN, UK
| | - Bethany M. James
- School of Biosciences, The University of Sheffield, Sheffield S10 2TN, UK
- Neuroscience Institute, The University of Sheffield, Sheffield S10 2TN, UK
| | - Daniel Bose
- School of Biosciences, The University of Sheffield, Sheffield S10 2TN, UK
- Neuroscience Institute, The University of Sheffield, Sheffield S10 2TN, UK
| | - Jacqueline Kim Dale
- Division of Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee DD1 4HN, UK
| | - Anestis Tsakiridis
- School of Biosciences, The University of Sheffield, Sheffield S10 2TN, UK
- Neuroscience Institute, The University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
17
|
Liu C, Xie Y, Chen X, Liu L, Liu C, Yin Z. BAF45D-binding to HOX genes was differentially targeted in H9-derived spinal cord neural stem cells. Sci Rep 2024; 14:29. [PMID: 38168763 PMCID: PMC10761701 DOI: 10.1038/s41598-023-50939-y] [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: 06/28/2023] [Accepted: 12/28/2023] [Indexed: 01/05/2024] Open
Abstract
Chromatin accessibility has been used to define how cells adopt region-specific neural fates. BAF45D is one of the subunits of a specialised chromatin remodelling BAF complex. It has been reported that BAF45D is expressed in spinal cord neural stem cells (NSCs) and regulates their fate specification. Within the developing vertebrate spinal cord, HOX genes exhibit spatially restricted expression patterns. However, the chromatin accessibility of BAF45D binding HOX genes in spinal cord NSCs is unclear. In the present study, we found that in H9-derived spinal cord NSCs, BAF45D targets TBX6, a gene that regulates spinal cord neural mesodermal progenitors. Furthermore, BAF45D binding to the NES gene is much more enriched in H9-derived spinal cord NSCs chromatin compared to ESCs chromatin. In addition, BAF45D binding to anterior and trunk/central HOX genes, but not to lumbosacral HOX genes, was much more enriched in NSCs chromatin compared to ESCs chromatin. These results may shed new light on the role of BAF45D in regulating region-specific spinal cord NSCs by targeting HOX genes.
Collapse
Affiliation(s)
- Chang Liu
- Department of Orthopedics, The First Affiliated Hospital, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Yuxin Xie
- Department of Histology and Embryology, Institute of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Xueying Chen
- Department of Histology and Embryology, Institute of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Lihua Liu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Chao Liu
- Department of Histology and Embryology, Institute of Stem Cell and Tissue Engineering, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, Anhui, China.
| | - Zongsheng Yin
- Department of Orthopedics, The First Affiliated Hospital, Anhui Medical University, Hefei, 230032, Anhui, China.
| |
Collapse
|
18
|
Miller A, Dasen JS. Establishing and maintaining Hox profiles during spinal cord development. Semin Cell Dev Biol 2024; 152-153:44-57. [PMID: 37029058 PMCID: PMC10524138 DOI: 10.1016/j.semcdb.2023.03.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/18/2023] [Accepted: 03/30/2023] [Indexed: 04/09/2023]
Abstract
The chromosomally-arrayed Hox gene family plays central roles in embryonic patterning and the specification of cell identities throughout the animal kingdom. In vertebrates, the relatively large number of Hox genes and pervasive expression throughout the body has hindered understanding of their biological roles during differentiation. Studies on the subtype diversification of spinal motor neurons (MNs) have provided a tractable system to explore the function of Hox genes during differentiation, and have provided an entry point to explore how neuronal fate determinants contribute to motor circuit assembly. Recent work, using both in vitro and in vivo models of MN subtype differentiation, have revealed how patterning morphogens and regulation of chromatin structure determine cell-type specific programs of gene expression. These studies have not only shed light on basic mechanisms of rostrocaudal patterning in vertebrates, but also have illuminated mechanistic principles of gene regulation that likely operate in the development and maintenance of terminal fates in other systems.
Collapse
Affiliation(s)
- Alexander Miller
- NYU Neuroscience Institute and Developmental Genetics Programs, Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY 10016, USA.
| | - Jeremy S Dasen
- NYU Neuroscience Institute and Developmental Genetics Programs, Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
19
|
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
|
20
|
Masak G, Davidson LA. Constructing the pharyngula: Connecting the primary axial tissues of the head with the posterior axial tissues of the tail. Cells Dev 2023; 176:203866. [PMID: 37394035 PMCID: PMC10756936 DOI: 10.1016/j.cdev.2023.203866] [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: 03/23/2023] [Revised: 06/04/2023] [Accepted: 06/29/2023] [Indexed: 07/04/2023]
Abstract
The pharyngula stage of vertebrate development is characterized by stereotypical arrangement of ectoderm, mesoderm, and neural tissues from the anterior spinal cord to the posterior, yet unformed tail. While early embryologists over-emphasized the similarity between vertebrate embryos at the pharyngula stage, there is clearly a common architecture upon which subsequent developmental programs generate diverse cranial structures and epithelial appendages such as fins, limbs, gills, and tails. The pharyngula stage is preceded by two morphogenetic events: gastrulation and neurulation, which establish common shared structures despite the occurrence of cellular processes that are distinct to each of the species. Even along the body axis of a singular organism, structures with seemingly uniform phenotypic characteristics at the pharyngula stage have been established by different processes. We focus our review on the processes underlying integration of posterior axial tissue formation with the primary axial tissues that creates the structures laid out in the pharyngula. Single cell sequencing and novel gene targeting technologies have provided us with new insights into the differences between the processes that form the anterior and posterior axis, but it is still unclear how these processes are integrated to create a seamless body. We suggest that the primary and posterior axial tissues in vertebrates form through distinct mechanisms and that the transition between these mechanisms occur at different locations along the anterior-posterior axis. Filling gaps that remain in our understanding of this transition could resolve ongoing problems in organoid culture and regeneration.
Collapse
Affiliation(s)
- Geneva Masak
- Integrative Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Lance A Davidson
- Integrative Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA; Department of Developmental Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA; Department of Computational and Systems Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| |
Collapse
|
21
|
Ong ALC, Kokaji T, Kishi A, Takihara Y, Shinozuka T, Shimamoto R, Isotani A, Shirai M, Sasai N. Acquisition of neural fate by combination of BMP blockade and chromatin modification. iScience 2023; 26:107887. [PMID: 37771660 PMCID: PMC10522999 DOI: 10.1016/j.isci.2023.107887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/07/2023] [Accepted: 09/07/2023] [Indexed: 09/30/2023] Open
Abstract
Neural induction is a process where naive cells are converted into committed cells with neural characteristics, and it occurs at the earliest step during embryogenesis. Although the signaling molecules and chromatin remodeling for neural induction have been identified, the mutual relationships between these molecules are yet to be fully understood. By taking advantage of the neural differentiation system of mouse embryonic stem (ES) cells, we discovered that the BMP signal regulates the expression of several polycomb repressor complex (PRC) component genes. We particularly focused on Polyhomeotic Homolog 1 (Phc1) and established Phc1-knockout (Phc1-KO) ES cells. We found that Phc1-KO failed to acquire the neural fate, and the cells remained in pluripotent or primitive non-neural states. Chromatin accessibility analysis suggests that Phc1 is essential for chromatin packing. Aberrant upregulation of the BMP signal was confirmed in the Phc1 homozygotic mutant embryos. Taken together, Phc1 is required for neural differentiation through epigenetic modification.
Collapse
Affiliation(s)
- Agnes Lee Chen Ong
- Division of Biological Sciences, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma 630-0192, Japan
| | - Toshiya Kokaji
- Data-driven biology, NAIST Data Science Center, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma 630-0192, Japan
| | - Arisa Kishi
- Division of Biological Sciences, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma 630-0192, Japan
| | - Yoshihiro Takihara
- Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima 734-0037, Japan
| | - Takuma Shinozuka
- Division of Biological Sciences, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma 630-0192, Japan
| | - Ren Shimamoto
- Division of Biological Sciences, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma 630-0192, Japan
| | - Ayako Isotani
- Division of Biological Sciences, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma 630-0192, Japan
| | - Manabu Shirai
- Omics Research Center (ORC), National Cerebral and Cardiovascular Center, 6-1 Kishibe Shinmachi, Suita, Osaka 564-8565, Japan
| | - Noriaki Sasai
- Division of Biological Sciences, Nara Institute of Science and Technology, 8916-5 Takayama-cho, Ikoma 630-0192, Japan
| |
Collapse
|
22
|
Bohic M, Upadhyay A, Eisdorfer JT, Keating J, Simon RC, Briones BA, Azadegan C, Nacht HD, Oputa O, Martinez AM, Bethell BN, Gradwell MA, Romanienko P, Ramer MS, Stuber GD, Abraira VE. A new Hoxb8FlpO mouse line for intersectional approaches to dissect developmentally defined adult sensorimotor circuits. Front Mol Neurosci 2023; 16:1176823. [PMID: 37603775 PMCID: PMC10437123 DOI: 10.3389/fnmol.2023.1176823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 07/04/2023] [Indexed: 08/23/2023] Open
Abstract
Improvements in the speed and cost of expression profiling of neuronal tissues offer an unprecedented opportunity to define ever finer subgroups of neurons for functional studies. In the spinal cord, single cell RNA sequencing studies support decades of work on spinal cord lineage studies, offering a unique opportunity to probe adult function based on developmental lineage. While Cre/Flp recombinase intersectional strategies remain a powerful tool to manipulate spinal neurons, the field lacks genetic tools and strategies to restrict manipulations to the adult mouse spinal cord at the speed at which new tools develop. This study establishes a new workflow for intersectional mouse-viral strategies to dissect adult spinal function based on developmental lineages in a modular fashion. To restrict manipulations to the spinal cord, we generate a brain-sparing Hoxb8FlpO mouse line restricting Flp recombinase expression to caudal tissue. Recapitulating endogenous Hoxb8 gene expression, Flp-dependent reporter expression is present in the caudal embryo starting day 9.5. This expression restricts Flp activity in the adult to the caudal brainstem and below. Hoxb8FlpO heterozygous and homozygous mice do not develop any of the sensory or locomotor phenotypes evident in Hoxb8 heterozygous or mutant animals, suggesting normal developmental function of the Hoxb8 gene and protein in Hoxb8FlpO mice. Compared to the variability of brain recombination in available caudal Cre and Flp lines, Hoxb8FlpO activity is not present in the brain above the caudal brainstem, independent of mouse genetic background. Lastly, we combine the Hoxb8FlpO mouse line with dorsal horn developmental lineage Cre mouse lines to express GFP in developmentally determined dorsal horn populations. Using GFP-dependent Cre recombinase viruses and Cre recombinase-dependent inhibitory chemogenetics, we target developmentally defined lineages in the adult. We show how developmental knock-out versus transient adult silencing of the same ROR𝛃 lineage neurons affects adult sensorimotor behavior. In summary, this new mouse line and viral approach provides a blueprint to dissect adult somatosensory circuit function using Cre/Flp genetic tools to target spinal cord interneurons based on genetic lineage.
Collapse
Affiliation(s)
- Manon Bohic
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Aman Upadhyay
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- Neuroscience PhD Program at Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - Jaclyn T. Eisdorfer
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Jessica Keating
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- School of Medicine, Oregon Health and Science University, Portland, OR, United States
- M.D./PhD Program in Neuroscience, School of Medicine, Oregon Health and Science University, Portland, OR, United States
| | - Rhiana C. Simon
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Brandy A. Briones
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Chloe Azadegan
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Hannah D. Nacht
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Olisemeka Oputa
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Alana M. Martinez
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Bridget N. Bethell
- International Collaboration on Repair Discoveries and Department of Zoology, The University of British Columbia, Vancouver, BC, Canada
| | - Mark A. Gradwell
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| | - Peter Romanienko
- Genome Editing Shared Resource, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Matt S. Ramer
- International Collaboration on Repair Discoveries and Department of Zoology, The University of British Columbia, Vancouver, BC, Canada
| | - Garret D. Stuber
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Victoria E. Abraira
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, Piscataway, NJ, United States
| |
Collapse
|
23
|
Sen SQ. Generating neural diversity through spatial and temporal patterning. Semin Cell Dev Biol 2023; 142:54-66. [PMID: 35738966 DOI: 10.1016/j.semcdb.2022.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 06/08/2022] [Accepted: 06/08/2022] [Indexed: 11/19/2022]
Abstract
The nervous system consists of a vast diversity of neurons and glia that are accurately assembled into functional circuits. What are the mechanisms that generate these diverse cell types? During development, an epithelial sheet with neurogenic potential is initially regionalised into spatially restricted domains of gene expression. From this, pools of neural stem cells (NSCs) with distinct molecular profiles and the potential to generate different neuron types, are specified. These NSCs then divide asymmetrically to self-renew and generate post-mitotic neurons or glia. As NSCs age, they experience transitions in gene expression, which further allows them to generate different neurons or glia over time. Versions of this general template of spatial and temporal patterning operate during the development of different parts of different nervous systems. Here, I cover our current knowledge of Drosophila brain and optic lobe development as well as the development of the vertebrate cortex and spinal cord within the framework of this above template. I highlight where our knowledge is lacking, where mechanisms beyond these might operate, and how the emergence of new technologies might help address unanswered questions.
Collapse
Affiliation(s)
- Sonia Q Sen
- Tata Institute for Genetics and Society, UAS-GKVK Campus, Bellary Road, Bangalore, India.
| |
Collapse
|
24
|
Buchner F, Dokuzluoglu Z, Grass T, Rodriguez-Muela N. Spinal Cord Organoids to Study Motor Neuron Development and Disease. Life (Basel) 2023; 13:1254. [PMID: 37374039 PMCID: PMC10303776 DOI: 10.3390/life13061254] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 05/18/2023] [Indexed: 06/29/2023] Open
Abstract
Motor neuron diseases (MNDs) are a heterogeneous group of disorders that affect the cranial and/or spinal motor neurons (spMNs), spinal sensory neurons and the muscular system. Although they have been investigated for decades, we still lack a comprehensive understanding of the underlying molecular mechanisms; and therefore, efficacious therapies are scarce. Model organisms and relatively simple two-dimensional cell culture systems have been instrumental in our current knowledge of neuromuscular disease pathology; however, in the recent years, human 3D in vitro models have transformed the disease-modeling landscape. While cerebral organoids have been pursued the most, interest in spinal cord organoids (SCOs) is now also increasing. Pluripotent stem cell (PSC)-based protocols to generate SpC-like structures, sometimes including the adjacent mesoderm and derived skeletal muscle, are constantly being refined and applied to study early human neuromuscular development and disease. In this review, we outline the evolution of human PSC-derived models for generating spMN and recapitulating SpC development. We also discuss how these models have been applied to exploring the basis of human neurodevelopmental and neurodegenerative diseases. Finally, we provide an overview of the main challenges to overcome in order to generate more physiologically relevant human SpC models and propose some exciting new perspectives.
Collapse
Affiliation(s)
- Felix Buchner
- German Center for Neurodegenerative Diseases, 01307 Dresden, Germany; (F.B.); (Z.D.); (T.G.)
| | - Zeynep Dokuzluoglu
- German Center for Neurodegenerative Diseases, 01307 Dresden, Germany; (F.B.); (Z.D.); (T.G.)
| | - Tobias Grass
- German Center for Neurodegenerative Diseases, 01307 Dresden, Germany; (F.B.); (Z.D.); (T.G.)
| | - Natalia Rodriguez-Muela
- German Center for Neurodegenerative Diseases, 01307 Dresden, Germany; (F.B.); (Z.D.); (T.G.)
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, 01307 Dresden, Germany
- Max Planck Institute for Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| |
Collapse
|
25
|
Sullivan AE. Epigenetic Control of Cell Potency and Fate Determination during Mammalian Gastrulation. Genes (Basel) 2023; 14:1143. [PMID: 37372324 PMCID: PMC10298296 DOI: 10.3390/genes14061143] [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: 04/21/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
Pluripotent embryonic stem cells have a unique and characteristic epigenetic profile, which is critical for differentiation to all embryonic germ lineages. When stem cells exit the pluripotent state and commit to lineage-specific identities during the process of gastrulation in early embryogenesis, extensive epigenetic remodelling mediates both the switch in cellular programme and the loss of potential to adopt alternative lineage programmes. However, it remains to be understood how the stem cell epigenetic profile encodes pluripotency, or how dynamic epigenetic regulation helps to direct cell fate specification. Recent advances in stem cell culture techniques, cellular reprogramming, and single-cell technologies that can quantitatively profile epigenetic marks have led to significant insights into these questions, which are important for understanding both embryonic development and cell fate engineering. This review provides an overview of key concepts and highlights exciting new advances in the field.
Collapse
Affiliation(s)
- Adrienne E. Sullivan
- Quantitative Stem Cell Biology Lab, Francis Crick Institute, London NW1 1AT, UK;
- Adelaide Centre for Epigenetics, School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide 5000, Australia
- South Australian immunoGENomics Cancer Institute (SAiGENCI), Faculty of Health and Medical Sciences, University of Adelaide, Adelaide 5000, Australia
| |
Collapse
|
26
|
Schuster HC, Hirth F. Phylogenetic tracing of midbrain-specific regulatory sequences suggests single origin of eubilaterian brains. SCIENCE ADVANCES 2023; 9:eade8259. [PMID: 37224241 PMCID: PMC10208574 DOI: 10.1126/sciadv.ade8259] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 04/18/2023] [Indexed: 05/26/2023]
Abstract
Conserved cis-regulatory elements (CREs) control Engrailed-, Pax2-, and dachshund-related gene expression networks directing the formation and function of corresponding midbrain circuits in arthropods and vertebrates. Polarized outgroup analyses of 31 sequenced metazoan genomes representing all animal clades reveal the emergence of Pax2- and dachshund-related CRE-like sequences in anthozoan Cnidaria. The full complement, including Engrailed-related CRE-like sequences, is only detectable in spiralians, ecdysozoans, and chordates that have a brain; they exhibit comparable genomic locations and extensive nucleotide identities that reveal the presence of a conserved core domain, all of which are absent in non-neural genes and, together, distinguish them from randomly assembled sequences. Their presence concurs with a genetic boundary separating the rostral from caudal nervous systems, demonstrated for the metameric brains of annelids, arthropods, and chordates and the asegmental cycloneuralian and urochordate brain. These findings suggest that gene regulatory networks for midbrain circuit formation evolved within the lineage that led to the common ancestor of protostomes and deuterostomes.
Collapse
Affiliation(s)
- Helen C. Schuster
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, and Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | | |
Collapse
|
27
|
Blair JD, Hartman A, Zenk F, Dalgarno C, Treutlein B, Satija R. Phospho-seq: Integrated, multi-modal profiling of intracellular protein dynamics in single cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.27.534442. [PMID: 37034703 PMCID: PMC10081255 DOI: 10.1101/2023.03.27.534442] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/17/2023]
Abstract
Cell signaling plays a critical role in regulating cellular behavior and fate. While multimodal single-cell sequencing technologies are rapidly advancing, scalable and flexible profiling of cell signaling states alongside other molecular modalities remains challenging. Here we present Phospho-seq, an integrated approach that aims to quantify phosphorylated intracellular and intranuclear proteins, and to connect their activity with cis-regulatory elements and transcriptional targets. We utilize a simplified benchtop antibody conjugation method to create large custom antibody panels for simultaneous protein and scATAC-seq profiling on whole cells, and integrate this information with scRNA-seq datasets via bridge integration. We apply our workflow to cell lines, induced pluripotent stem cells, and 3-month-old brain organoids to demonstrate its broad applicability. We demonstrate that Phospho-seq can define cellular states and trajectories, reconstruct gene regulatory relationships, and characterize the causes and consequences of heterogeneous cell signaling in neurodevelopment.
Collapse
Affiliation(s)
- John D. Blair
- New York Genome Center, New York, NY
- New York University, Center for Genomics and Systems Biology, New York, NY
| | | | | | | | | | - Rahul Satija
- New York Genome Center, New York, NY
- New York University, Center for Genomics and Systems Biology, New York, NY
| |
Collapse
|
28
|
Xu HJ, Yao Y, Yao F, Chen J, Li M, Yang X, Li S, Lu F, Hu P, He S, Peng G, Jing N. Generation of functional posterior spinal motor neurons from hPSCs-derived human spinal cord neural progenitor cells. CELL REGENERATION (LONDON, ENGLAND) 2023; 12:15. [PMID: 36949352 PMCID: PMC10033800 DOI: 10.1186/s13619-023-00159-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 02/24/2023] [Indexed: 03/24/2023]
Abstract
Spinal motor neurons deficiency results in a series of devastating disorders such as amyotrophic lateral sclerosis (ALS), spinal muscular atrophy (SMA) and spinal cord injury (SCI). These disorders are currently incurable, while human pluripotent stem cells (hPSCs)-derived spinal motor neurons are promising but suffered from inappropriate regional identity and functional immaturity for the study and treatment of posterior spinal cord related injuries. In this study, we have established human spinal cord neural progenitor cells (hSCNPCs) via hPSCs differentiated neuromesodermal progenitors (NMPs) and demonstrated the hSCNPCs can be continuously expanded up to 40 passages. hSCNPCs can be rapidly differentiated into posterior spinal motor neurons with high efficiency. The functional maturity has been examined in detail. Moreover, a co-culture scheme which is compatible for both neural and muscular differentiation is developed to mimic the neuromuscular junction (NMJ) formation in vitro. Together, these studies highlight the potential avenues for generating clinically relevant spinal motor neurons and modeling neuromuscular diseases through our defined hSCNPCs.
Collapse
Affiliation(s)
- He Jax Xu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yao Yao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Fenyong Yao
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Jiehui Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Meishi Li
- University of Chinese Academy of Sciences, Beijing, China
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Xianfa Yang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, China
- Guangzhou Laboratory/Bioland Laboratory, Guangzhou, 510005, China
| | - Sheng Li
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
- Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 20023, China
| | - Fangru Lu
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Ping Hu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China
- University of Chinese Academy of Sciences, Beijing, China
- Guangzhou Laboratory/Bioland Laboratory, Guangzhou, 510005, China
- Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 20023, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
| | - Shuijin He
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| | - Guangdun Peng
- University of Chinese Academy of Sciences, Beijing, China.
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Center for Cell Lineage and Atlas, Bioland Laboratory, Guangzhou, 510005, China.
| | - Naihe Jing
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Guangzhou Laboratory/Bioland Laboratory, Guangzhou, 510005, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
29
|
Ahmad MH, Ghosh B, Rizvi MA, Ali M, Kaur L, Mondal AC. Neural crest cells development and neuroblastoma progression: Role of Wnt signaling. J Cell Physiol 2023; 238:306-328. [PMID: 36502519 DOI: 10.1002/jcp.30931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 09/19/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022]
Abstract
Neuroblastoma (NB) is one of the most common heterogeneous extracranial cancers in infancy that arises from neural crest (NC) cells of the sympathetic nervous system. The Wnt signaling pathway, both canonical and noncanonical pathway, is a highly conserved signaling pathway that regulates the development and differentiation of the NC cells during embryogenesis. Reports suggest that aberrant activation of Wnt ligands/receptors in Wnt signaling pathways promote progression and relapse of NB. Wnt signaling pathways regulate NC induction and migration in a similar manner; it regulates proliferation and metastasis of NB. Inhibiting the Wnt signaling pathway or its ligands/receptors induces apoptosis and abrogates proliferation and tumorigenicity in all major types of NB cells. Here, we comprehensively discuss the Wnt signaling pathway and its mechanisms in regulating the development of NC and NB pathogenesis. This review highlights the implications of aberrant Wnt signaling in the context of etiology, progression, and relapse of NB. We have also described emerging strategies for Wnt-based therapies against the progression of NB that will provide new insights into the development of Wnt-based therapeutic strategies for NB.
Collapse
Affiliation(s)
- Mir Hilal Ahmad
- School of Life Sciences, Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India.,Genome Biology Lab, Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Balaram Ghosh
- Department of Clinical Pharmacology, Midnapore Medical College & Hospital, West Bengal, Medinipur, India
| | - Moshahid Alam Rizvi
- Genome Biology Lab, Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Mansoor Ali
- School of Life Sciences, Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Loveleena Kaur
- Division of Cancer Pharmacology, Indian Institute of Integrative Medicine (IIIM), Srinagar, India
| | - Amal Chandra Mondal
- School of Life Sciences, Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
30
|
Delás MJ, Kalaitzis CM, Fawzi T, Demuth M, Zhang I, Stuart HT, Costantini E, Ivanovitch K, Tanaka EM, Briscoe J. Developmental cell fate choice in neural tube progenitors employs two distinct cis-regulatory strategies. Dev Cell 2023; 58:3-17.e8. [PMID: 36516856 PMCID: PMC7614300 DOI: 10.1016/j.devcel.2022.11.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 10/10/2022] [Accepted: 11/22/2022] [Indexed: 12/15/2022]
Abstract
In many developing tissues, the patterns of gene expression that assign cell fate are organized by graded secreted signals. Cis-regulatory elements (CREs) interpret these signals to control gene expression, but how this is accomplished remains poorly understood. In the neural tube, a gradient of the morphogen sonic hedgehog (Shh) patterns neural progenitors. We identify two distinct ways in which CREs translate graded Shh into differential gene expression in mouse neural progenitors. In most progenitors, a common set of CREs control gene activity by integrating cell-type-specific inputs. By contrast, the most ventral progenitors use a unique set of CREs, established by the pioneer factor FOXA2. This parallels the role of FOXA2 in endoderm, where FOXA2 binds some of the same sites. Together, the data identify distinct cis-regulatory strategies for the interpretation of morphogen signaling and raise the possibility of an evolutionarily conserved role for FOXA2 across tissues.
Collapse
Affiliation(s)
| | | | - Tamara Fawzi
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | | | - Isabel Zhang
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Hannah T Stuart
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), 1030 Vienna, Austria
| | - Elena Costantini
- Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), 1030 Vienna, Austria
| | | | - Elly M Tanaka
- Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), 1030 Vienna, Austria
| | - James Briscoe
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
31
|
Cable J, Lutolf MP, Fu J, Park SE, Apostolou A, Chen S, Song CJ, Spence JR, Liberali P, Lancaster M, Meier AB, Pek NMQ, Wells JM, Capeling MM, Uzquiano A, Musah S, Huch M, Gouti M, Hombrink P, Quadrato G, Urenda JP. Organoids as tools for fundamental discovery and translation-a Keystone Symposia report. Ann N Y Acad Sci 2022; 1518:196-208. [PMID: 36177906 PMCID: PMC11293861 DOI: 10.1111/nyas.14874] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Complex three-dimensional in vitro organ-like models, or organoids, offer a unique biological tool with distinct advantages over two-dimensional cell culture systems, which can be too simplistic, and animal models, which can be too complex and may fail to recapitulate human physiology and pathology. Significant progress has been made in driving stem cells to differentiate into different organoid types, though several challenges remain. For example, many organoid models suffer from high heterogeneity, and it can be difficult to fully incorporate the complexity of in vivo tissue and organ development to faithfully reproduce human biology. Successfully addressing such limitations would increase the viability of organoids as models for drug development and preclinical testing. On April 3-6, 2022, experts in organoid development and biology convened at the Keystone Symposium "Organoids as Tools for Fundamental Discovery and Translation" to discuss recent advances and insights from this relatively new model system into human development and disease.
Collapse
Affiliation(s)
| | - Matthias P Lutolf
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Lausanne, Switzerland
- Institute of Chemical Sciences and Engineering, School of Basic Science (SB), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Roche Institute for Translational Bioengineering (ITB), Pharma Research and Early Development (pRED), F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
- Department of Cell & Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sunghee Estelle Park
- Department of Bioengineering and NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Athanasia Apostolou
- Emulate Inc, Boston, Massachusetts, USA
- Department of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medical College, New York City, New York, USA
| | - Cheng Jack Song
- Keck Medicine of University of Southern California, Los Angeles, California, USA
| | - Jason R Spence
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Prisca Liberali
- Friedrich Miescher Institute for Biomedical Research (FMI) and University of Basel, Basel, Switzerland
| | | | - Anna B Meier
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Nicole Min Qian Pek
- Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati, Ohio, USA
- Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - James M Wells
- Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati, Ohio, USA
- Division of Developmental Biology and Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Meghan M Capeling
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan, USA
| | - Ana Uzquiano
- Department of Stem Cell and Regenerative Biology, Harvard University
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Samira Musah
- Developmental and Stem Cell Biology Program and Division of Nephrology, Department of Medicine and Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina, USA
- Center for Biomolecular and Tissue Engineering, Durham, North Carolina, USA
- Department of Biomedical Engineering, Pratt School of Engineering, Durham, North Carolina, USA
- Duke Regeneration Center, Duke University, Durham, North Carolina, USA
| | - Meritxell Huch
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Mina Gouti
- Stem Cell Modelling of Development & Disease Group, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Pleun Hombrink
- University Medical Center Utrecht and HUB Organoids, Utrecht, Netherlands
| | - Giorgia Quadrato
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine and Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, California, USA
| | - Jean-Paul Urenda
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine and Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
32
|
Semprich CI, Davidson L, Amorim Torres A, Patel H, Briscoe J, Metzis V, Storey KG. ERK1/2 signalling dynamics promote neural differentiation by regulating chromatin accessibility and the polycomb repressive complex. PLoS Biol 2022; 20:e3000221. [PMID: 36455041 PMCID: PMC9746999 DOI: 10.1371/journal.pbio.3000221] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 12/13/2022] [Accepted: 10/11/2022] [Indexed: 12/05/2022] Open
Abstract
Fibroblast growth factor (FGF) is a neural inducer in many vertebrate embryos, but how it regulates chromatin organization to coordinate the activation of neural genes is unclear. Moreover, for differentiation to progress, FGF signalling must decline. Why these signalling dynamics are required has not been determined. Here, we show that dephosphorylation of the FGF effector kinase ERK1/2 rapidly increases chromatin accessibility at neural genes in mouse embryos, and, using ATAC-seq in human embryonic stem cell derived spinal cord precursors, we demonstrate that this occurs genome-wide across neural genes. Importantly, ERK1/2 inhibition induces precocious neural gene transcription, and this involves dissociation of the polycomb repressive complex from key gene loci. This takes place independently of subsequent loss of the repressive histone mark H3K27me3 and transcriptional onset. Transient ERK1/2 inhibition is sufficient for the dissociation of the repressive complex, and this is not reversed on resumption of ERK1/2 signalling. Moreover, genomic footprinting of sites identified by ATAC-seq together with ChIP-seq for polycomb protein Ring1B revealed that ERK1/2 inhibition promotes the occupancy of neural transcription factors (TFs) at non-polycomb as well as polycomb associated sites. Together, these findings indicate that ERK1/2 signalling decline promotes global changes in chromatin accessibility and TF binding at neural genes by directing polycomb and other regulators and appears to serve as a gating mechanism that provides directionality to the process of differentiation.
Collapse
Affiliation(s)
- Claudia I. Semprich
- Division of Cell & Developmental Biology, School of Life Sciences, University of Dundee, Scotland, United Kingdom
| | - Lindsay Davidson
- Division of Cell & Developmental Biology, School of Life Sciences, University of Dundee, Scotland, United Kingdom
| | - Adriana Amorim Torres
- Division of Cell & Developmental Biology, School of Life Sciences, University of Dundee, Scotland, United Kingdom
| | | | | | - Vicki Metzis
- The Francis Crick Institute, London, United Kingdom
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
- * E-mail: (VM); (KGS)
| | - Kate G. Storey
- Division of Cell & Developmental Biology, School of Life Sciences, University of Dundee, Scotland, United Kingdom
- * E-mail: (VM); (KGS)
| |
Collapse
|
33
|
Diacou R, Nandigrami P, Fiser A, Liu W, Ashery-Padan R, Cvekl A. Cell fate decisions, transcription factors and signaling during early retinal development. Prog Retin Eye Res 2022; 91:101093. [PMID: 35817658 PMCID: PMC9669153 DOI: 10.1016/j.preteyeres.2022.101093] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 12/30/2022]
Abstract
The development of the vertebrate eyes is a complex process starting from anterior-posterior and dorso-ventral patterning of the anterior neural tube, resulting in the formation of the eye field. Symmetrical separation of the eye field at the anterior neural plate is followed by two symmetrical evaginations to generate a pair of optic vesicles. Next, reciprocal invagination of the optic vesicles with surface ectoderm-derived lens placodes generates double-layered optic cups. The inner and outer layers of the optic cups develop into the neural retina and retinal pigment epithelium (RPE), respectively. In vitro produced retinal tissues, called retinal organoids, are formed from human pluripotent stem cells, mimicking major steps of retinal differentiation in vivo. This review article summarizes recent progress in our understanding of early eye development, focusing on the formation the eye field, optic vesicles, and early optic cups. Recent single-cell transcriptomic studies are integrated with classical in vivo genetic and functional studies to uncover a range of cellular mechanisms underlying early eye development. The functions of signal transduction pathways and lineage-specific DNA-binding transcription factors are dissected to explain cell-specific regulatory mechanisms underlying cell fate determination during early eye development. The functions of homeodomain (HD) transcription factors Otx2, Pax6, Lhx2, Six3 and Six6, which are required for early eye development, are discussed in detail. Comprehensive understanding of the mechanisms of early eye development provides insight into the molecular and cellular basis of developmental ocular anomalies, such as optic cup coloboma. Lastly, modeling human development and inherited retinal diseases using stem cell-derived retinal organoids generates opportunities to discover novel therapies for retinal diseases.
Collapse
Affiliation(s)
- Raven Diacou
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Prithviraj Nandigrami
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Andras Fiser
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Wei Liu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Ruth Ashery-Padan
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Ales Cvekl
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
34
|
Elder N, Fattahi F, McDevitt TC, Zholudeva LV. Diseased, differentiated and difficult: Strategies for improved engineering of in vitro neurological systems. Front Cell Neurosci 2022; 16:962103. [PMID: 36238834 PMCID: PMC9550918 DOI: 10.3389/fncel.2022.962103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 08/22/2022] [Indexed: 12/01/2022] Open
Abstract
The rapidly growing field of cellular engineering is enabling scientists to more effectively create in vitro models of disease and develop specific cell types that can be used to repair damaged tissue. In particular, the engineering of neurons and other components of the nervous system is at the forefront of this field. The methods used to engineer neural cells can be largely divided into systems that undergo directed differentiation through exogenous stimulation (i.e., via small molecules, arguably following developmental pathways) and those that undergo induced differentiation via protein overexpression (i.e., genetically induced and activated; arguably bypassing developmental pathways). Here, we highlight the differences between directed differentiation and induced differentiation strategies, how they can complement one another to generate specific cell phenotypes, and impacts of each strategy on downstream applications. Continued research in this nascent field will lead to the development of improved models of neurological circuits and novel treatments for those living with neurological injury and disease.
Collapse
Affiliation(s)
- Nicholas Elder
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, United States
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States
- Gladstone Institutes, San Francisco, CA, United States
| | - Faranak Fattahi
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, United States
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States
| | - Todd C. McDevitt
- Gladstone Institutes, San Francisco, CA, United States
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, United States
- Sana Biotechnology, Inc., South San Francisco, CA, United States
| | - Lyandysha V. Zholudeva
- Gladstone Institutes, San Francisco, CA, United States
- *Correspondence: Lyandysha V. Zholudeva,
| |
Collapse
|
35
|
Bolkhovitinov L, Weselman BT, Shaw GA, Dong C, Giribhattanavar J, Saha MS. Tissue Rotation of the Xenopus Anterior-Posterior Neural Axis Reveals Profound but Transient Plasticity at the Mid-Gastrula Stage. J Dev Biol 2022; 10:38. [PMID: 36135371 PMCID: PMC9503425 DOI: 10.3390/jdb10030038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/02/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022] Open
Abstract
The establishment of anterior-posterior (AP) regional identity is an essential step in the appropriate development of the vertebrate central nervous system. An important aspect of AP neural axis formation is the inherent plasticity that allows developing cells to respond to and recover from the various perturbations that embryos continually face during the course of development. While the mechanisms governing the regionalization of the nervous system have been extensively studied, relatively less is known about the nature and limits of early neural plasticity of the anterior-posterior neural axis. This study aims to characterize the degree of neural axis plasticity in Xenopus laevis by investigating the response of embryos to a 180-degree rotation of their AP neural axis during gastrula stages by assessing the expression of regional marker genes using in situ hybridization. Our results reveal the presence of a narrow window of time between the mid- and late gastrula stage, during which embryos are able undergo significant recovery following a 180-degree rotation of their neural axis and eventually express appropriate regional marker genes including Otx, Engrailed, and Krox. By the late gastrula stage, embryos show misregulation of regional marker genes following neural axis rotation, suggesting that this profound axial plasticity is a transient phenomenon that is lost by late gastrula stages.
Collapse
Affiliation(s)
- Lyuba Bolkhovitinov
- Department of Molecular Biology, Massachusetts General Hospital, Harvard University, Boston, MA 02114, USA
| | - Bryan T. Weselman
- School of Medicine, Georgetown University, Washington, DC 20007, USA
| | - Gladys A. Shaw
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Chen Dong
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Margaret S. Saha
- Department of Biology, College of William and Mary, Williamsburg, VA 23185, USA
| |
Collapse
|
36
|
Iyer NR, Ashton RS. Bioengineering the human spinal cord. Front Cell Dev Biol 2022; 10:942742. [PMID: 36092702 PMCID: PMC9458954 DOI: 10.3389/fcell.2022.942742] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/01/2022] [Indexed: 12/04/2022] Open
Abstract
Three dimensional, self-assembled organoids that recapitulate key developmental and organizational events during embryogenesis have proven transformative for the study of human central nervous system (CNS) development, evolution, and disease pathology. Brain organoids have predominated the field, but human pluripotent stem cell (hPSC)-derived models of the spinal cord are on the rise. This has required piecing together the complex interactions between rostrocaudal patterning, which specifies axial diversity, and dorsoventral patterning, which establishes locomotor and somatosensory phenotypes. Here, we review how recent insights into neurodevelopmental biology have driven advancements in spinal organoid research, generating experimental models that have the potential to deepen our understanding of neural circuit development, central pattern generation (CPG), and neurodegenerative disease along the body axis. In addition, we discuss the application of bioengineering strategies to drive spinal tissue morphogenesis in vitro, current limitations, and future perspectives on these emerging model systems.
Collapse
Affiliation(s)
- Nisha R. Iyer
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
- Wisconsin Institute for Discovery, University of Wisconsin—Madison, Madison, WI, United States
- Department of Biomedical Engineering, University of Wisconsin—Madison, Madison, WI, United States
| | - Randolph S. Ashton
- Wisconsin Institute for Discovery, University of Wisconsin—Madison, Madison, WI, United States
- Department of Biomedical Engineering, University of Wisconsin—Madison, Madison, WI, United States
| |
Collapse
|
37
|
Ávila-González D, Portillo W, Barragán-Álvarez CP, Hernandez-Montes G, Flores-Garza E, Molina-Hernández A, Diaz-Martinez NE, Diaz NF. The human amniotic epithelium confers a bias to differentiate toward the neuroectoderm lineage in human embryonic stem cells. eLife 2022; 11:68035. [PMID: 35815953 PMCID: PMC9313526 DOI: 10.7554/elife.68035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 07/08/2022] [Indexed: 11/28/2022] Open
Abstract
Human embryonic stem cells (hESCs) derive from the epiblast and have pluripotent potential. To maintain the conventional conditions of the pluripotent potential in an undifferentiated state, inactivated mouse embryonic fibroblast (iMEF) is used as a feeder layer. However, it has been suggested that hESC under this conventional condition (hESC-iMEF) is an artifact that does not correspond to the in vitro counterpart of the human epiblast. Our previous studies demonstrated the use of an alternative feeder layer of human amniotic epithelial cells (hAECs) to derive and maintain hESC. We wondered if the hESC-hAEC culture could represent a different pluripotent stage than that of naïve or primed conventional conditions, simulating the stage in which the amniotic epithelium derives from the epiblast during peri-implantation. Like the conventional primed hESC-iMEF, hESC-hAEC has the same levels of expression as the ‘pluripotency core’ and does not express markers of naïve pluripotency. However, it presents a downregulation of HOX genes and genes associated with the endoderm and mesoderm, and it exhibits an increase in the expression of ectoderm lineage genes, specifically in the anterior neuroectoderm. Transcriptome analysis showed in hESC-hAEC an upregulated signature of genes coding for transcription factors involved in neural induction and forebrain development, and the ability to differentiate into a neural lineage was superior in comparison with conventional hESC-iMEF. We propose that the interaction of hESC with hAEC confers hESC a biased potential that resembles the anteriorized epiblast, which is predisposed to form the neural ectoderm.
Collapse
Affiliation(s)
- Daniela Ávila-González
- Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City, Mexico
| | - Wendy Portillo
- Behavioral and Cognitive Neurobiology, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Carla P Barragán-Álvarez
- Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco, Guadalajara, Mexico
| | | | - Eliezer Flores-Garza
- Departamento de Biología Molecular y Biotecnología, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Anayansi Molina-Hernández
- Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City, Mexico
| | | | - Nestor F Diaz
- Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City, Mexico
| |
Collapse
|
38
|
Martin BL. Mesoderm induction and patterning: Insights from neuromesodermal progenitors. Semin Cell Dev Biol 2022; 127:37-45. [PMID: 34840081 PMCID: PMC9130346 DOI: 10.1016/j.semcdb.2021.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/02/2021] [Accepted: 11/10/2021] [Indexed: 12/23/2022]
Abstract
The discovery of mesoderm inducing signals helped usher in the era of molecular developmental biology, and today the mechanisms of mesoderm induction and patterning are still intensely studied. Mesoderm induction begins during gastrulation, but recent evidence in vertebrates shows that this process continues after gastrulation in a group of posteriorly localized cells called neuromesodermal progenitors (NMPs). NMPs reside within the post-gastrulation embryonic structure called the tailbud, where they make a lineage decision between ectoderm (spinal cord) and mesoderm. The majority of NMP-derived mesoderm generates somites, but also contributes to lateral mesoderm fates such as endothelium. The discovery of NMPs provides a new paradigm in which to study vertebrate mesoderm induction. This review will discuss mechanisms of mesoderm induction within NMPs, and how they have informed our understanding of mesoderm induction more broadly within vertebrates as well as animal species outside of the vertebrate lineage. Special focus will be given to the signaling networks underlying NMP-derived mesoderm induction and patterning, as well as emerging work on the significance of partial epithelial-mesenchymal states in coordinating cell fate and morphogenesis.
Collapse
Affiliation(s)
- Benjamin L Martin
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA.
| |
Collapse
|
39
|
Cooper F, Tsakiridis A. Towards clinical applications of in vitro-derived axial progenitors. Dev Biol 2022; 489:110-117. [PMID: 35718236 DOI: 10.1016/j.ydbio.2022.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/28/2022] [Accepted: 06/14/2022] [Indexed: 11/19/2022]
Abstract
The production of the tissues that make up the mammalian embryonic trunk takes place in a head-tail direction, via the differentiation of posteriorly-located axial progenitor populations. These include bipotent neuromesodermal progenitors (NMPs), which generate both spinal cord neurectoderm and presomitic mesoderm, the precursor of the musculoskeleton. Over the past few years, a number of studies have described the derivation of NMP-like cells from mouse and human pluripotent stem cells (PSCs). In turn, these have greatly facilitated the establishment of PSC differentiation protocols aiming to give rise efficiently to posterior mesodermal and neural cell types, which have been particularly challenging to produce using previous approaches. Moreover, the advent of 3-dimensional-based culture systems incorporating distinct axial progenitor-derived cell lineages has opened new avenues toward the functional dissection of early patterning events and cell vs non-cell autonomous effects. Here, we provide a brief overview of the applications of these cell types in disease modelling and cell therapy and speculate on their potential uses in the future.
Collapse
Affiliation(s)
- Fay Cooper
- Centre for Stem Cell Biology, School of Bioscience, The University of Sheffield, Western Bank, Sheffield, S10 2TN, United Kingdom; Neuroscience Institute, The University of Sheffield, Western Bank, Sheffield, S10 2TN, United Kingdom
| | - Anestis Tsakiridis
- Centre for Stem Cell Biology, School of Bioscience, The University of Sheffield, Western Bank, Sheffield, S10 2TN, United Kingdom; Neuroscience Institute, The University of Sheffield, Western Bank, Sheffield, S10 2TN, United Kingdom.
| |
Collapse
|
40
|
Investigating chromatin accessibility during development and differentiation by ATAC-sequencing to guide the identification of cis-regulatory elements. Biochem Soc Trans 2022; 50:1167-1177. [PMID: 35604124 PMCID: PMC9246326 DOI: 10.1042/bst20210834] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 11/17/2022]
Abstract
Mapping accessible chromatin across time scales can give insights into its dynamic nature, for example during cellular differentiation and tissue or organism development. Analysis of such data can be utilised to identify functional cis-regulatory elements (CRE) and transcription factor binding sites and, when combined with transcriptomics, can reveal gene regulatory networks (GRNs) of expressed genes. Chromatin accessibility mapping is a powerful approach and can be performed using ATAC-sequencing (ATAC-seq), whereby Tn5 transposase inserts sequencing adaptors into genomic DNA to identify differentially accessible regions of chromatin in different cell populations. It requires low sample input and can be performed and analysed relatively quickly compared with other methods. The data generated from ATAC-seq, along with other genomic approaches, can help uncover chromatin packaging and potential cis-regulatory elements that may be responsible for gene expression. Here, we describe the ATAC-seq approach and give examples from mainly vertebrate embryonic development, where such datasets have identified the highly dynamic nature of chromatin, with differing landscapes between cellular precursors for different lineages.
Collapse
|
41
|
Blassberg R, Patel H, Watson T, Gouti M, Metzis V, Delás MJ, Briscoe J. Sox2 levels regulate the chromatin occupancy of WNT mediators in epiblast progenitors responsible for vertebrate body formation. Nat Cell Biol 2022; 24:633-644. [PMID: 35550614 PMCID: PMC9106585 DOI: 10.1038/s41556-022-00910-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/29/2022] [Indexed: 02/07/2023]
Abstract
WNT signalling has multiple roles. It maintains pluripotency of embryonic stem cells, assigns posterior identity in the epiblast and induces mesodermal tissue. Here we provide evidence that these distinct functions are conducted by the transcription factor SOX2, which adopts different modes of chromatin interaction and regulatory element selection depending on its level of expression. At high levels, SOX2 displaces nucleosomes from regulatory elements with high-affinity SOX2 binding sites, recruiting the WNT effector TCF/β-catenin and maintaining pluripotent gene expression. Reducing SOX2 levels destabilizes pluripotency and reconfigures SOX2/TCF/β-catenin occupancy to caudal epiblast expressed genes. These contain low-affinity SOX2 sites and are co-occupied by T/Bra and CDX. The loss of SOX2 allows WNT-induced mesodermal differentiation. These findings define a role for Sox2 levels in dictating the chromatin occupancy of TCF/β-catenin and reveal how context-specific responses to a signal are configured by the level of a transcription factor.
Collapse
Affiliation(s)
| | | | | | - Mina Gouti
- Stem Cell Modelling of Development & Disease Group, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Vicki Metzis
- The Francis Crick Institute, London, UK
- Institute of Clinical Sciences, Imperial College London, London, UK
| | | | | |
Collapse
|
42
|
Sears KE, Gullapalli K, Trivedi D, Mihas A, Bukys MA, Jensen J. Controlling neural territory patterning from pluripotency using a systems developmental biology approach. iScience 2022; 25:104133. [PMID: 35434550 PMCID: PMC9010746 DOI: 10.1016/j.isci.2022.104133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 06/09/2021] [Accepted: 03/17/2022] [Indexed: 11/18/2022] Open
Abstract
Successful manufacture of specialized human cells requires process understanding of directed differentiation. Here, we apply high-dimensional Design of Experiments (HD-DoE) methodology to identify critical process parameters (CPPs) that govern neural territory patterning from pluripotency—the first stage toward specification of central nervous system (CNS) cell fates. Using computerized experimental design, 7 developmental signaling pathways were simultaneously perturbed in human pluripotent stem cell culture. Regionally specific genes spanning the anterior-posterior and dorsal-ventral axes of the developing embryo were measured after 3 days and mathematical models describing pathway control were developed using regression analysis. High-dimensional models revealed particular combinations of signaling inputs that induce expression profiles consistent with emerging CNS territories and defined CPPs for anterior and posterior neuroectoderm patterning. The results demonstrate the importance of combinatorial control during neural induction and challenge the use of generic neural induction strategies such as dual-SMAD inhibition, when seeking to specify particular lineages from pluripotency. Mathematical models describe pathway control of neuroectoderm marker expression Stage 1 media conditions optimized for regionally specific neuroectoderm in 3 days Optimized conditions are more consistent than dual-SMADi across hiPSC lines
Collapse
|
43
|
Needham J, Metzis V. Heads or tails: Making the spinal cord. Dev Biol 2022; 485:80-92. [DOI: 10.1016/j.ydbio.2022.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/15/2021] [Accepted: 03/02/2022] [Indexed: 12/14/2022]
|
44
|
Sáez M, Blassberg R, Camacho-Aguilar E, Siggia ED, Rand DA, Briscoe J. Statistically derived geometrical landscapes capture principles of decision-making dynamics during cell fate transitions. Cell Syst 2022; 13:12-28.e3. [PMID: 34536382 PMCID: PMC8785827 DOI: 10.1016/j.cels.2021.08.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 07/06/2021] [Accepted: 08/23/2021] [Indexed: 12/20/2022]
Abstract
Fate decisions in developing tissues involve cells transitioning between discrete cell states, each defined by distinct gene expression profiles. The Waddington landscape, in which the development of a cell is viewed as a ball rolling through a valley filled terrain, is an appealing way to describe differentiation. To construct and validate accurate landscapes, quantitative methods based on experimental data are necessary. We combined principled statistical methods with a framework based on catastrophe theory and approximate Bayesian computation to formulate a quantitative dynamical landscape that accurately predicts cell fate outcomes of pluripotent stem cells exposed to different combinations of signaling factors. Analysis of the landscape revealed two distinct ways in which cells make a binary choice between one of two fates. We suggest that these represent archetypal designs for developmental decisions. The approach is broadly applicable for the quantitative analysis of differentiation and for determining the logic of developmental decisions.
Collapse
Affiliation(s)
- Meritxell Sáez
- Mathematics Institute, University of Warwick, Coventry CV4 7AL, UK; The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; Zeeman Institute for Systems Biology and Infectious Epidemiology Research, University of Warwick, Coventry CV4 7AL, UK
| | | | - Elena Camacho-Aguilar
- Mathematics Institute, University of Warwick, Coventry CV4 7AL, UK; Department of Biosciences, Rice University, Houston, TX 77005, USA
| | - Eric D Siggia
- Center for Studies of Physics and Biology, the Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - David A Rand
- Mathematics Institute, University of Warwick, Coventry CV4 7AL, UK; Zeeman Institute for Systems Biology and Infectious Epidemiology Research, University of Warwick, Coventry CV4 7AL, UK.
| | - James Briscoe
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
45
|
Yan Y, Wang Q. BMP Signaling: Lighting up the Way for Embryonic Dorsoventral Patterning. Front Cell Dev Biol 2022; 9:799772. [PMID: 35036406 PMCID: PMC8753366 DOI: 10.3389/fcell.2021.799772] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/06/2021] [Indexed: 11/13/2022] Open
Abstract
One of the most significant events during early embryonic development is the establishment of a basic embryonic body plan, which is defined by anteroposterior, dorsoventral (DV), and left-right axes. It is well-known that the morphogen gradient created by BMP signaling activity is crucial for DV axis patterning across a diverse set of vertebrates. The regulation of BMP signaling during DV patterning has been strongly conserved across evolution. This is a remarkable regulatory and evolutionary feat, as the BMP gradient has been maintained despite the tremendous variation in embryonic size and shape across species. Interestingly, the embryonic DV axis exhibits robust stability, even in face of variations in BMP signaling. Multiple lines of genetic, molecular, and embryological evidence have suggested that numerous BMP signaling components and their attendant regulators act in concert to shape the developing DV axis. In this review, we summarize the current knowledge of the function and regulation of BMP signaling in DV patterning. Throughout, we focus specifically on popular model animals, such as Xenopus and zebrafish, highlighting the similarities and differences of the regulatory networks between species. We also review recent advances regarding the molecular nature of DV patterning, including the initiation of the DV axis, the formation of the BMP gradient, and the regulatory molecular mechanisms behind BMP signaling during the establishment of the DV axis. Collectively, this review will help clarify our current understanding of the molecular nature of DV axis formation.
Collapse
Affiliation(s)
- Yifang Yan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.,National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Qiang Wang
- State Key Laboratory of Membrane Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
46
|
Sawai A, Pfennig S, Bulajić M, Miller A, Khodadadi-Jamayran A, Mazzoni EO, Dasen JS. PRC1 sustains the integrity of neural fate in the absence of PRC2 function. eLife 2022; 11:e72769. [PMID: 34994686 PMCID: PMC8765755 DOI: 10.7554/elife.72769] [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: 08/04/2021] [Accepted: 01/06/2022] [Indexed: 12/13/2022] Open
Abstract
Polycomb repressive complexes (PRCs) 1 and 2 maintain stable cellular memories of early fate decisions by establishing heritable patterns of gene repression. PRCs repress transcription through histone modifications and chromatin compaction, but their roles in neuronal subtype diversification are poorly defined. We found that PRC1 is essential for the specification of segmentally restricted spinal motor neuron (MN) subtypes, while PRC2 activity is dispensable to maintain MN positional identities during terminal differentiation. Mutation of the core PRC1 component Ring1 in mice leads to increased chromatin accessibility and ectopic expression of a broad variety of fates determinants, including Hox transcription factors, while neuronal class-specific features are maintained. Loss of MN subtype identities in Ring1 mutants is due to the suppression of Hox-dependent specification programs by derepressed Hox13 paralogs (Hoxa13, Hoxb13, Hoxc13, Hoxd13). These results indicate that PRC1 can function in the absence of de novo PRC2-dependent histone methylation to maintain chromatin topology and postmitotic neuronal fate.
Collapse
Affiliation(s)
- Ayana Sawai
- Neuroscience Institute, Department of Neuroscience and Physiology, NYU School of MedicineNew YorkUnited States
| | - Sarah Pfennig
- Neuroscience Institute, Department of Neuroscience and Physiology, NYU School of MedicineNew YorkUnited States
| | - Milica Bulajić
- Department of Biology, New York UniversityNew YorkUnited States
| | - Alexander Miller
- Neuroscience Institute, Department of Neuroscience and Physiology, NYU School of MedicineNew YorkUnited States
| | - Alireza Khodadadi-Jamayran
- Applied Bioinformatics Laboratories, Office of Science and Research, NYU School of MedcineNew YorkUnited States
| | | | - Jeremy S Dasen
- Neuroscience Institute, Department of Neuroscience and Physiology, NYU School of MedicineNew YorkUnited States
| |
Collapse
|
47
|
Dasen JS. Establishing the Molecular and Functional Diversity of Spinal Motoneurons. ADVANCES IN NEUROBIOLOGY 2022; 28:3-44. [PMID: 36066819 DOI: 10.1007/978-3-031-07167-6_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Spinal motoneurons are a remarkably diverse class of neurons responsible for facilitating a broad range of motor behaviors and autonomic functions. Studies of motoneuron differentiation have provided fundamental insights into the developmental mechanisms of neuronal diversification, and have illuminated principles of neural fate specification that operate throughout the central nervous system. Because of their relative anatomical simplicity and accessibility, motoneurons have provided a tractable model system to address multiple facets of neural development, including early patterning, neuronal migration, axon guidance, and synaptic specificity. Beyond their roles in providing direct communication between central circuits and muscle, recent studies have revealed that motoneuron subtype-specific programs also play important roles in determining the central connectivity and function of motor circuits. Cross-species comparative analyses have provided novel insights into how evolutionary changes in subtype specification programs may have contributed to adaptive changes in locomotor behaviors. This chapter focusses on the gene regulatory networks governing spinal motoneuron specification, and how studies of spinal motoneurons have informed our understanding of the basic mechanisms of neuronal specification and spinal circuit assembly.
Collapse
Affiliation(s)
- Jeremy S Dasen
- NYU Neuroscience Institute, Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY, USA.
| |
Collapse
|
48
|
Binagui-Casas A, Dias A, Guillot C, Metzis V, Saunders D. Building consensus in neuromesodermal research: Current advances and future biomedical perspectives. Curr Opin Cell Biol 2021; 73:133-140. [PMID: 34717142 DOI: 10.1016/j.ceb.2021.08.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 08/12/2021] [Accepted: 08/31/2021] [Indexed: 10/20/2022]
Abstract
The development of the vertebrate body axis relies on the activity of different populations of axial progenitors, including neuromesodermal progenitors. Currently, the term 'Neuromesodermal progenitors' is associated with various definitions. Here, we use distinct terminologies to highlight advances in our understanding of this cell type at both the single-cell and population levels. We discuss how these recent insights prompt new opportunities to address a range of biomedical questions spanning cancer metastasis, congenital disorders, cellular metabolism, regenerative medicine, and evolution. Finally, we outline some of the major unanswered questions and propose future directions at the forefront of neuromesodermal research.
Collapse
Affiliation(s)
- Anahí Binagui-Casas
- Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - André Dias
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, 2780-156, Oeiras, Portugal.
| | - Charlène Guillot
- Department of Pathology, Brigham and Women's Hospital & Department of Genetics, Harvard Medical School, 60 Fenwood Road, Boston, MA, USA; Institute of Genetics, Reproduction and Development, Medical school, University of Clermont Auvergne, 28, Place Henri Dunant, 63001 Clermont-Ferrand, France
| | - Vicki Metzis
- Institute of Clinical Sciences, Imperial College London, London, W12 0NN, UK
| | - Dillan Saunders
- Department of Genetics, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK
| |
Collapse
|
49
|
Sagner A, Zhang I, Watson T, Lazaro J, Melchionda M, Briscoe J. A shared transcriptional code orchestrates temporal patterning of the central nervous system. PLoS Biol 2021; 19:e3001450. [PMID: 34767545 PMCID: PMC8612522 DOI: 10.1371/journal.pbio.3001450] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 11/24/2021] [Accepted: 10/20/2021] [Indexed: 01/13/2023] Open
Abstract
The molecular mechanisms that produce the full array of neuronal subtypes in the vertebrate nervous system are incompletely understood. Here, we provide evidence of a global temporal patterning program comprising sets of transcription factors that stratifies neurons based on the developmental time at which they are generated. This transcriptional code acts throughout the central nervous system, in parallel to spatial patterning, thereby increasing the diversity of neurons generated along the neuraxis. We further demonstrate that this temporal program operates in stem cell-derived neurons and is under the control of the TGFβ signaling pathway. Targeted perturbation of components of the temporal program, Nfia and Nfib, reveals their functional requirement for the generation of late-born neuronal subtypes. Together, our results provide evidence for the existence of a previously unappreciated global temporal transcriptional program of neuronal subtype identity and suggest that the integration of spatial and temporal patterning mechanisms diversifies and organizes neuronal subtypes in the vertebrate nervous system.
Collapse
Affiliation(s)
- Andreas Sagner
- The Francis Crick Institute, London, United Kingdom
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Isabel Zhang
- The Francis Crick Institute, London, United Kingdom
| | | | - Jorge Lazaro
- The Francis Crick Institute, London, United Kingdom
| | | | | |
Collapse
|
50
|
Zheng Y, Zhang F, Xu S, Wu L. Advances in neural organoid systems and their application in neurotoxicity testing of environmental chemicals. Genes Environ 2021; 43:39. [PMID: 34551827 PMCID: PMC8456188 DOI: 10.1186/s41021-021-00214-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/05/2021] [Indexed: 12/15/2022] Open
Abstract
Due to the complex structure and function of central nervous system (CNS), human CNS in vitro modeling is still a great challenge. Neurotoxicity testing of environmental chemicals mainly depends on the traditional animal models, which have various limitations such as species differences, expensive and time-consuming. Meanwhile, in vitro two-dimensional (2D) cultured cells or three-dimensional (3D) cultured neurospheres cannot fully simulate complex 3D structure of neural tissues. Recent advancements in neural organoid systems provides excellent models for the testing of environmental chemicals that affect the development of human CNS. Neural organoids derived from hPSCs not only can simulate the process of CNS development, including early stage neural tube formation, neuroepithelium differentiation and regional specification, but also its 3D structure, thus can be used to evaluate the effect of chemicals on differentiation and morphogenesis. Here, we provide a review of recent progress in the methods of culturing neural organoids and their applications in neurotoxicity testing of environmental chemicals. We conclude by highlighting challenge and future directions in neurotoxicity testing based on neural organoids.
Collapse
Affiliation(s)
- Yuanyuan Zheng
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, China
| | - Fangrong Zhang
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, China
| | - Shengmin Xu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, China
| | - Lijun Wu
- Information Materials and Intelligent Sensing Laboratory of Anhui Province, Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, China.
| |
Collapse
|