101
|
Taguchi A, Nishinakamura R. Higher-Order Kidney Organogenesis from Pluripotent Stem Cells. Cell Stem Cell 2017; 21:730-746.e6. [PMID: 29129523 DOI: 10.1016/j.stem.2017.10.011] [Citation(s) in RCA: 274] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 08/27/2017] [Accepted: 10/20/2017] [Indexed: 11/30/2022]
Abstract
Organogenesis generates higher-order structures containing functional subunits, connective components, and progenitor niches. Despite recent advances in organoid-based modeling of tissue development, recapitulating these complex configurations from pluripotent stem cells (PSCs) has remained challenging. In this study, we report assembly of kidney organoids that recapitulate embryonic branching morphogenesis. By studying the distinct origins and developmental processes of the ureteric bud, which contains epithelial kidney progenitors that undergo branching morphogenesis and thereby plays a central role in orchestrating organ geometry, and neighboring mesenchymal nephron progenitors, we established a protocol for differential induction of each lineage from mouse and human PSCs. Importantly, reassembled organoids developed the inherent architectures of the embryonic kidney, including the peripheral progenitor niche and internally differentiated nephrons that were interconnected by a ramified ureteric epithelium. This selective induction and reassembly strategy will be a powerful approach to recapitulate organotypic architecture in PSC-derived organoids.
Collapse
Affiliation(s)
- Atsuhiro Taguchi
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Kumamoto 860-0811, Japan.
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Kumamoto 860-0811, Japan.
| |
Collapse
|
102
|
Das D, Chatti V, Emonet T, Holley SA. Patterned Disordered Cell Motion Ensures Vertebral Column Symmetry. Dev Cell 2017; 42:170-180.e5. [PMID: 28743003 DOI: 10.1016/j.devcel.2017.06.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 03/30/2017] [Accepted: 06/23/2017] [Indexed: 12/24/2022]
Abstract
The biomechanics of posterior embryonic growth must be dynamically regulated to ensure bilateral symmetry of the spinal column. Throughout vertebrate trunk elongation, motile mesodermal progenitors undergo an order-to-disorder transition via an epithelial-to-mesenchymal transition and sort symmetrically into the left and right paraxial mesoderm. We combine theoretical modeling of cell migration in a tail-bud-like geometry with experimental data analysis to assess the importance of ordered and disordered cell motion. We find that increasing order in cell motion causes a phase transition from symmetric to asymmetric body elongation. In silico and in vivo, overly ordered cell motion converts normal anisotropic fluxes into stable vortices near the posterior tail bud, contributing to asymmetric cell sorting. Thus, disorder is a physical mechanism that ensures the bilateral symmetry of the spinal column. These physical properties of the tissue connect across scales such that patterned disorder at the cellular level leads to the emergence of organism-level order.
Collapse
Affiliation(s)
- Dipjyoti Das
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Veena Chatti
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Thierry Emonet
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA; Department of Physics, Yale University, New Haven, CT, USA.
| | - Scott A Holley
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA.
| |
Collapse
|
103
|
Antagonistic Activities of Sox2 and Brachyury Control the Fate Choice of Neuro-Mesodermal Progenitors. Dev Cell 2017; 42:514-526.e7. [DOI: 10.1016/j.devcel.2017.07.021] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 06/07/2017] [Accepted: 07/24/2017] [Indexed: 12/25/2022]
|
104
|
Neijts R, Deschamps J. At the base of colinear Hox gene expression: cis -features and trans -factors orchestrating the initial phase of Hox cluster activation. Dev Biol 2017; 428:293-299. [DOI: 10.1016/j.ydbio.2017.02.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/16/2017] [Indexed: 10/19/2022]
|
105
|
Sharma R, Shafer MER, Bareke E, Tremblay M, Majewski J, Bouchard M. Bmp signaling maintains a mesoderm progenitor cell state in the mouse tailbud. Development 2017; 144:2982-2993. [PMID: 28705896 DOI: 10.1242/dev.149955] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 07/10/2017] [Indexed: 02/01/2023]
Abstract
Caudal somites are generated from a pool of progenitor cells located in the tailbud region. These progenitor cells form the presomitic mesoderm that gradually differentiates into somites under the action of the segmentation clock. The signals responsible for tailbud mesoderm progenitor pool maintenance during axial elongation are still elusive. Here, we show that Bmp signaling is sufficient to activate the entire mesoderm progenitor gene signature in primary cultures of caudal mesoderm cells. Bmp signaling acts through the key regulatory genes brachyury (T) and Nkx1-2 and contributes to the activation of several other regulators of the mesoderm progenitor gene network. In the absence of Bmp signaling, tailbud mesoderm progenitor cells acquire aberrant gene expression signatures of the heart, blood, muscle and skeletal embryonic lineages. Treatment of embryos with the Bmp inhibitor noggin confirmed the requirement for Bmp signaling for normal T expression and the prevention of abnormal lineage marker activation. Together, these results identify Bmp signaling as a non-cell-autonomous signal necessary for mesoderm progenitor cell homeostasis.
Collapse
Affiliation(s)
- Richa Sharma
- Goodman Cancer Research Centre and Department of Biochemistry, McGill University, Montreal, Canada H3A 1A3
| | - Maxwell E R Shafer
- Goodman Cancer Research Centre and Department of Biochemistry, McGill University, Montreal, Canada H3A 1A3
| | - Eric Bareke
- Department of Human Genetics, McGill University and Genome Quebec Innovation Centre, Montreal, Canada H3A 0G1
| | - Mathieu Tremblay
- Goodman Cancer Research Centre and Department of Biochemistry, McGill University, Montreal, Canada H3A 1A3
| | - Jacek Majewski
- Department of Human Genetics, McGill University and Genome Quebec Innovation Centre, Montreal, Canada H3A 0G1
| | - Maxime Bouchard
- Goodman Cancer Research Centre and Department of Biochemistry, McGill University, Montreal, Canada H3A 1A3
| |
Collapse
|
106
|
Abstract
During vertebrate embryonic development, the spinal cord is formed by the neural derivatives of a neuromesodermal population that is specified at early stages of development and which develops in concert with the caudal regression of the primitive streak. Several processes related to spinal cord specification and maturation are coupled to this caudal extension including neurogenesis, ventral patterning and neural crest specification and all of them seem to be crucially regulated by Fibroblast Growth Factor (FGF) signaling, which is prominently active in the neuromesodermal region and transiently in its derivatives. Here we review the role of FGF signaling in those processes, trying to separate its different functions and highlighting the interactions with other signaling pathways. Finally, these early functions of FGF signaling in spinal cord development may underlay partly its ability to promote regeneration in the lesioned spinal cord as well as its action promoting specific fates in neural stem cell cultures that may be used for therapeutical purposes.
Collapse
Affiliation(s)
- Ruth Diez Del Corral
- Department of Cellular, Molecular and Developmental Neurobiology, Cajal Institute, Consejo Superior de Investigaciones CientíficasMadrid, Spain.,Champalimaud Research, Champalimaud Centre for the UnknownLisbon, Portugal
| | - Aixa V Morales
- Department of Cellular, Molecular and Developmental Neurobiology, Cajal Institute, Consejo Superior de Investigaciones CientíficasMadrid, Spain
| |
Collapse
|
107
|
Liao BK, Oates AC. Delta-Notch signalling in segmentation. ARTHROPOD STRUCTURE & DEVELOPMENT 2017; 46:429-447. [PMID: 27888167 PMCID: PMC5446262 DOI: 10.1016/j.asd.2016.11.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 11/20/2016] [Accepted: 11/21/2016] [Indexed: 06/06/2023]
Abstract
Modular body organization is found widely across multicellular organisms, and some of them form repetitive modular structures via the process of segmentation. It's vastly interesting to understand how these regularly repeated structures are robustly generated from the underlying noise in biomolecular interactions. Recent studies from arthropods reveal similarities in segmentation mechanisms with vertebrates, and raise the possibility that the three phylogenetic clades, annelids, arthropods and chordates, might share homology in this process from a bilaterian ancestor. Here, we discuss vertebrate segmentation with particular emphasis on the role of the Notch intercellular signalling pathway. We introduce vertebrate segmentation and Notch signalling, pointing out historical milestones, then describe existing models for the Notch pathway in the synchronization of noisy neighbouring oscillators, and a new role in the modulation of gene expression wave patterns. We ask what functions Notch signalling may have in arthropod segmentation and explore the relationship between Notch-mediated lateral inhibition and synchronization. Finally, we propose open questions and technical challenges to guide future investigations into Notch signalling in segmentation.
Collapse
Affiliation(s)
- Bo-Kai Liao
- Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, London NW7 1AA, UK
| | - Andrew C Oates
- Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, London NW7 1AA, UK; Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
108
|
Gouti M, Delile J, Stamataki D, Wymeersch FJ, Huang Y, Kleinjung J, Wilson V, Briscoe J. A Gene Regulatory Network Balances Neural and Mesoderm Specification during Vertebrate Trunk Development. Dev Cell 2017; 41:243-261.e7. [PMID: 28457792 PMCID: PMC5425255 DOI: 10.1016/j.devcel.2017.04.002] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 02/20/2017] [Accepted: 04/03/2017] [Indexed: 01/02/2023]
Abstract
Transcriptional networks, regulated by extracellular signals, control cell fate decisions and determine the size and composition of developing tissues. One example is the network controlling bipotent neuromesodermal progenitors (NMPs) that fuel embryo elongation by generating spinal cord and trunk mesoderm tissue. Here, we use single-cell transcriptomics to identify the molecular signature of NMPs and reverse engineer the mechanism that regulates their differentiation. Together with genetic perturbations, this reveals a transcriptional network that integrates opposing retinoic acid (RA) and Wnt signals to determine the rate at which cells enter and exit the NMP state. RA, produced by newly generated mesodermal cells, provides feedback that initiates NMP generation and induces neural differentiation, thereby coordinating the production of neural and mesodermal tissue. Together, the data define a regulatory network architecture that balances the generation of different cell types from bipotential progenitors in order to facilitate orderly axis elongation. Single-cell RNA-seq reveals a signature of neuromesodermal progenitors In vitro NMPs resemble and differentiate similar to their in vivo counterparts Dual role for retinoic acid signaling in NMP induction and neural differentiation A transcriptional network regulates neural versus mesodermal allocation
Collapse
Affiliation(s)
- Mina Gouti
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK; Max Delbrück Center for Molecular Medicine, Robert-Rössle-Strasse 10, 13125 Berlin, Germany.
| | - Julien Delile
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | | | - Filip J Wymeersch
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Yali Huang
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Jens Kleinjung
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Valerie Wilson
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - James Briscoe
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
| |
Collapse
|
109
|
Huang C, Su T, Xue Y, Cheng C, Lay FD, McKee RA, Li M, Vashisht A, Wohlschlegel J, Novitch BG, Plath K, Kurdistani SK, Carey M. Cbx3 maintains lineage specificity during neural differentiation. Genes Dev 2017; 31:241-246. [PMID: 28270516 PMCID: PMC5358721 DOI: 10.1101/gad.292169.116] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 01/27/2017] [Indexed: 02/05/2023]
Abstract
Chromobox homolog 3 (Cbx3/heterochromatin protein 1γ [HP1γ]) stimulates cell differentiation, but its mechanism is unknown. We found that Cbx3 binds to gene promoters upon differentiation of murine embryonic stem cells (ESCs) to neural progenitor cells (NPCs) and recruits the Mediator subunit Med26. RNAi knockdown of either Cbx3 or Med26 inhibits neural differentiation while up-regulating genes involved in mesodermal lineage decisions. Thus, Cbx3 and Med26 together ensure the fidelity of lineage specification by enhancing the expression of neural genes and down-regulating genes specific to alternative fates.
Collapse
Affiliation(s)
- Chengyang Huang
- Department of Biological Chemistry, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles California 90095, USA
- Department of Neurobiology, Shantou University Medical College, Shantou 515041, China
| | - Trent Su
- Department of Biological Chemistry, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles California 90095, USA
| | - Yong Xue
- Department of Biological Chemistry, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles California 90095, USA
| | - Chen Cheng
- Department of Biological Chemistry, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles California 90095, USA
| | - Fides D Lay
- Department of Molecular, Cell, and Developmental Biology, University of California at Los Angeles, Los Angeles, California 90095, USA
| | - Robin A McKee
- Department of Biological Chemistry, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles California 90095, USA
| | - Meiyang Li
- Department of Neurobiology, Shantou University Medical College, Shantou 515041, China
| | - Ajay Vashisht
- Department of Biological Chemistry, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles California 90095, USA
| | - James Wohlschlegel
- Department of Biological Chemistry, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles California 90095, USA
| | - Bennett G Novitch
- Department of Neurobiology, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles California 90095, USA
| | - Kathrin Plath
- Department of Biological Chemistry, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles California 90095, USA
| | - Siavash K Kurdistani
- Department of Biological Chemistry, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles California 90095, USA
| | - Michael Carey
- Department of Biological Chemistry, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles California 90095, USA
| |
Collapse
|
110
|
Taniguchi Y, Kurth T, Weiche S, Reichelt S, Tazaki A, Perike S, Kappert V, Epperlein HH. The posterior neural plate in axolotl gives rise to neural tube or turns anteriorly to form somites of the tail and posterior trunk. Dev Biol 2017; 422:155-170. [DOI: 10.1016/j.ydbio.2016.12.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/21/2016] [Accepted: 12/21/2016] [Indexed: 11/28/2022]
|
111
|
Induction of nephron progenitors and glomeruli from human pluripotent stem cells. Pediatr Nephrol 2017; 32:195-200. [PMID: 26868670 DOI: 10.1007/s00467-016-3339-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 01/08/2016] [Accepted: 01/27/2016] [Indexed: 01/29/2023]
Abstract
Studies of kidney regeneration using stem cells have progressed rapidly in recent years. Our group has developed a protocol to induce nephron progenitors from both mouse and human pluripotent stem cells which is based on a revised model of early stage kidney specification. The induced progenitors readily reconstitute three-dimensional nephron structures, including glomeruli and renal tubules, in vitro. We can further generate human induced pluripotent stem cells (iPSCs), in which nephrin-expressing glomerular podocytes are tagged with green fluorescent protein (GFP). The sorted GFP-positive cells retain the podocyte-specific molecular and structural features. Upon transplantation, mouse endothelial cells of the host animals are integrated into the human iPSC-derived glomeruli, and the podocytes show further maturation. Other laboratories have reported different protocols to induce nephron structures from human iPSCs in vitro. These findings will accelerate our understanding of kidney development and diseases in humans.
Collapse
|
112
|
Houston DW. Vertebrate Axial Patterning: From Egg to Asymmetry. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 953:209-306. [PMID: 27975274 PMCID: PMC6550305 DOI: 10.1007/978-3-319-46095-6_6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The emergence of the bilateral embryonic body axis from a symmetrical egg has been a long-standing question in developmental biology. Historical and modern experiments point to an initial symmetry-breaking event leading to localized Wnt and Nodal growth factor signaling and subsequent induction and formation of a self-regulating dorsal "organizer." This organizer forms at the site of notochord cell internalization and expresses primarily Bone Morphogenetic Protein (BMP) growth factor antagonists that establish a spatiotemporal gradient of BMP signaling across the embryo, directing initial cell differentiation and morphogenesis. Although the basics of this model have been known for some time, many of the molecular and cellular details have only recently been elucidated and the extent that these events remain conserved throughout vertebrate evolution remains unclear. This chapter summarizes historical perspectives as well as recent molecular and genetic advances regarding: (1) the mechanisms that regulate symmetry-breaking in the vertebrate egg and early embryo, (2) the pathways that are activated by these events, in particular the Wnt pathway, and the role of these pathways in the formation and function of the organizer, and (3) how these pathways also mediate anteroposterior patterning and axial morphogenesis. Emphasis is placed on comparative aspects of the egg-to-embryo transition across vertebrates and their evolution. The future prospects for work regarding self-organization and gene regulatory networks in the context of early axis formation are also discussed.
Collapse
Affiliation(s)
- Douglas W Houston
- Department of Biology, The University of Iowa, 257 BB, Iowa City, IA, 52242, USA.
| |
Collapse
|
113
|
Neijts R, Amin S, van Rooijen C, Deschamps J. Cdx is crucial for the timing mechanism driving colinear Hox activation and defines a trunk segment in the Hox cluster topology. Dev Biol 2016; 422:146-154. [PMID: 28041967 DOI: 10.1016/j.ydbio.2016.12.024] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 12/09/2016] [Accepted: 12/27/2016] [Indexed: 11/28/2022]
Abstract
Cdx and Hox transcription factors are important regulators of axial patterning and are required for tissue generation along the vertebrate body axis. Cdx genes have been demonstrated to act upstream of Hox genes in midgestation embryos. Here, we investigate the role of Cdx transcription factors in the gradual colinear activation of the Hox clusters. We found that Hox temporally colinear expression is severely affected in epiblast stem cells derived from Cdx null embryos. We demonstrate that after initiation of 3' Hox gene transcription, Cdx activity is crucial for H3K27ac deposition and for accessibility of cis-regulatory elements around the central - or 'trunk' - Hox genes. We thereby identify a Cdx-responsive segment of HoxA, immediately 5' to the recently defined regulatory domain orchestrating initial transcription of the first Hox gene. We propose that this partition of HoxA into a Wnt-driven 3' part and the newly found Cdx-dependent middle segment of the cluster, forms a structural fundament of Hox colinearity of expression. Subsequently to initial Wnt-induced activation of 3' Hox genes, Cdx transcription factors would act as crucial effectors for activating central Hox genes, until the last gene of the cluster arrests the process.
Collapse
Affiliation(s)
- Roel Neijts
- Hubrecht Institute, Developmental Biology and Stem Cell Research, Uppsalalaan 8, 3584 CT Utrecht, and UMC Utrecht, the Netherlands
| | - Shilu Amin
- Hubrecht Institute, Developmental Biology and Stem Cell Research, Uppsalalaan 8, 3584 CT Utrecht, and UMC Utrecht, the Netherlands
| | - Carina van Rooijen
- Hubrecht Institute, Developmental Biology and Stem Cell Research, Uppsalalaan 8, 3584 CT Utrecht, and UMC Utrecht, the Netherlands
| | - Jacqueline Deschamps
- Hubrecht Institute, Developmental Biology and Stem Cell Research, Uppsalalaan 8, 3584 CT Utrecht, and UMC Utrecht, the Netherlands.
| |
Collapse
|
114
|
Molina A, Pituello F. Playing with the cell cycle to build the spinal cord. Dev Biol 2016; 432:14-23. [PMID: 28034699 DOI: 10.1016/j.ydbio.2016.12.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 12/14/2016] [Accepted: 12/20/2016] [Indexed: 12/31/2022]
Abstract
A fundamental issue in nervous system development and homeostasis is to understand the mechanisms governing the balance between the maintenance of proliferating progenitors versus their differentiation into post-mitotic neurons. Accumulating data suggest that the cell cycle and core regulators of the cell cycle machinery play a major role in regulating this fine balance. Here, we focus on the interplay between the cell cycle and cellular and molecular events governing spinal cord development. We describe the existing links between the cell cycle and interkinetic nuclear migration (INM). We show how the different morphogens patterning the neural tube also regulate the cell cycle machinery to coordinate proliferation and patterning. We give examples of how cell cycle core regulators regulate transcriptionally, or post-transcriptionally, genes involved in controlling the maintenance versus the differentiation of neural progenitors. Finally, we describe the changes in cell cycle kinetics occurring during neural tube patterning and at the time of neuronal differentiation, and we discuss future research directions to better understand the role of the cell cycle in cell fate decisions.
Collapse
Affiliation(s)
- Angie Molina
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, France.
| | - Fabienne Pituello
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, France.
| |
Collapse
|
115
|
Cdx and T Brachyury Co-activate Growth Signaling in the Embryonic Axial Progenitor Niche. Cell Rep 2016; 17:3165-3177. [DOI: 10.1016/j.celrep.2016.11.069] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 10/26/2016] [Accepted: 11/18/2016] [Indexed: 12/30/2022] Open
|
116
|
Kumar S, Cunningham TJ, Duester G. Nuclear receptor corepressors Ncor1 and Ncor2 (Smrt) are required for retinoic acid-dependent repression of Fgf8 during somitogenesis. Dev Biol 2016; 418:204-215. [PMID: 27506116 PMCID: PMC5031541 DOI: 10.1016/j.ydbio.2016.08.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 06/30/2016] [Accepted: 08/05/2016] [Indexed: 10/21/2022]
Abstract
Retinoic acid (RA) repression of Fgf8 is required for many different aspects of organogenesis, however relatively little is known about how endogenous RA controls gene repression as opposed to gene activation. Here, we show that nuclear receptor corepressors NCOR1 and NCOR2 (SMRT) redundantly mediate the ability of RA to repress Fgf8. Ncor1;Ncor2 double mutants generated by CRISPR/Cas9 gene editing exhibited a small somite and distended heart phenotype similar to that of RA-deficient Raldh2-/- embryos, associated with increased Fgf8 expression and FGF signaling in caudal progenitors and heart progenitors. Embryo chromatin immunoprecipitation studies revealed that NCOR1/2 but not coactivators are recruited to the Fgf8 RA response element (RARE) in an RA-dependent manner, whereas coactivators but not NCOR1/2 are recruited RA-dependently to a RARE near Rarb that is activated by RA. CRISPR/Cas9-mediated genomic deletion of the Fgf8 RARE in mouse embryos often resulted in a small somite defect with Fgf8 derepression caudally, but no defect was observed in heart development or heart Fgf8 expression. This suggests the existence of another DNA element whose function overlaps with the Fgf8 RARE to mediate Fgf8 repression by RA and NCOR1/2. Our studies support a model in which NCOR1/2 mediates direct RA-dependent repression of Fgf8 in caudal progenitors in order to control somitogenesis.
Collapse
Affiliation(s)
- Sandeep Kumar
- Development, Aging, and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Thomas J Cunningham
- Development, Aging, and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Gregg Duester
- Development, Aging, and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
117
|
Kondoh H, Takada S, Takemoto T. Axial level-dependent molecular and cellular mechanisms underlying the genesis of the embryonic neural plate. Dev Growth Differ 2016; 58:427-36. [PMID: 27279156 DOI: 10.1111/dgd.12295] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 04/22/2016] [Accepted: 04/22/2016] [Indexed: 01/18/2023]
Abstract
The transcription factor gene Sox2, centrally involved in neural primordial regulation, is activated by many enhancers. During the early stages of embryonic development, Sox2 is regulated by the enhancers N2 and N1 in the anterior neural plate (ANP) and posterior neural plate (PNP), respectively. This differential use of the enhancers reflects distinct regulatory mechanisms underlying the genesis of ANP and PNP. The ANP develops directly from the epiblast, triggered by nodal signal inhibition, and via the combined action of TFs SOX2, OTX2, POU3F1, and ZIC2, which promotes the the ANP development and inhibits other cell lineages. In contrast, the PNP is derived from neuromesodermal bipotential axial stem cells that develop into the neural plate when Sox2 is activated by the N1 enhancer, whereas they develop into the paraxial mesoderm when the N1 enhancer is repressed by the action of TBX6. The axial stem cells are maintained by the activity of WNT3a and T (Brachyury). However, at axial levels more anterior to the 8th somites (cervical levels), the development of both the neural plate and somite proceeds in the absence of WNT3a, T, or TBX6. These observations indicate that distinct molecular and cellular mechanisms determine neural plate genesis based on the axial level, and contradict the classical concept of the term "neural induction," which assumes a pan-neural plate mechanism.
Collapse
Affiliation(s)
- Hisato Kondoh
- Faculty of Life Sciences, Kyoto Sangyo University, Motoyama, Kamigamo, Kita-ku, Kyoto, 603-8555, Japan
| | - Shinji Takada
- Okazaki Institute for Integrative Bioscience, National Institutes of Natural Sciences, Okazaki, Aichi, 444-8787, Japan
| | - Tatsuya Takemoto
- Fujii Memorial Institute of Medical Sciences, Tokushima University, 3-18-15 Kuramoto-cho, Tokushima, 770-8503, Japan
| |
Collapse
|
118
|
Sudheer S, Liu J, Marks M, Koch F, Anurin A, Scholze M, Senft AD, Wittler L, Macura K, Grote P, Herrmann BG. Different Concentrations of FGF Ligands, FGF2 or FGF8 Determine Distinct States of WNT-Induced Presomitic Mesoderm. Stem Cells 2016; 34:1790-800. [DOI: 10.1002/stem.2371] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 02/07/2016] [Accepted: 03/03/2016] [Indexed: 12/25/2022]
Affiliation(s)
- Smita Sudheer
- Department of Developmental Genetics; Max Planck Institute for Molecular Genetics; Berlin Germany
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, The University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU; United Kingdom
| | - Jinhua Liu
- Department of Developmental Genetics; Max Planck Institute for Molecular Genetics; Berlin Germany
| | - Matthias Marks
- Department of Developmental Genetics; Max Planck Institute for Molecular Genetics; Berlin Germany
| | - Frederic Koch
- Department of Developmental Genetics; Max Planck Institute for Molecular Genetics; Berlin Germany
| | - Anna Anurin
- Department of Developmental Genetics; Max Planck Institute for Molecular Genetics; Berlin Germany
- Department of Biology; Chemistry and Pharmacy, Free University Berlin; Berlin Germany
| | - Manuela Scholze
- Department of Developmental Genetics; Max Planck Institute for Molecular Genetics; Berlin Germany
| | - Anna Dorothea Senft
- Department of Developmental Genetics; Max Planck Institute for Molecular Genetics; Berlin Germany
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE; United Kingdom
| | - Lars Wittler
- Department of Developmental Genetics; Max Planck Institute for Molecular Genetics; Berlin Germany
| | - Karol Macura
- Department of Developmental Genetics; Max Planck Institute for Molecular Genetics; Berlin Germany
| | - Phillip Grote
- Department of Developmental Genetics; Max Planck Institute for Molecular Genetics; Berlin Germany
- Institute of Cardiovascular Regeneration, Center for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, 60590, Frankfurt am Main; Germany
| | - Bernhard G. Herrmann
- Department of Developmental Genetics; Max Planck Institute for Molecular Genetics; Berlin Germany
| |
Collapse
|
119
|
Cearns MD, Escuin S, Alexandre P, Greene NDE, Copp AJ. Microtubules, polarity and vertebrate neural tube morphogenesis. J Anat 2016; 229:63-74. [PMID: 27025884 DOI: 10.1111/joa.12468] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2016] [Indexed: 12/20/2022] Open
Abstract
Microtubules (MTs) are key cellular components, long known to participate in morphogenetic events that shape the developing embryo. However, the links between the cellular functions of MTs, their effects on cell shape and polarity, and their role in large-scale morphogenesis remain poorly understood. Here, these relationships were examined with respect to two strategies for generating the vertebrate neural tube: bending and closure of the mammalian neural plate; and cavitation of the teleost neural rod. The latter process has been compared with 'secondary' neurulation that generates the caudal spinal cord in mammals. MTs align along the apico-basal axis of the mammalian neuroepithelium early in neural tube closure, participating functionally in interkinetic nuclear migration, which indirectly impacts on cell shape. Whether MTs play other functional roles in mammalian neurulation remains unclear. In the zebrafish, MTs are important for defining the neural rod midline prior to its cavitation, both by localizing apical proteins at the tissue midline and by orienting cell division through a mirror-symmetric MT apparatus that helps to further define the medial localization of apical polarity proteins. Par proteins have been implicated in centrosome positioning in neuroepithelia as well as in the control of polarized morphogenetic movements in the neural rod. Understanding of MT functions during early nervous system development has so far been limited, partly by techniques that fail to distinguish 'cause' from 'effect'. Future developments will likely rely on novel ways to selectively impair MT function in order to investigate the roles they play.
Collapse
Affiliation(s)
- Michael D Cearns
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, London, UK
| | - Sarah Escuin
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, London, UK
| | - Paula Alexandre
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, London, UK
| | - Nicholas D E Greene
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, London, UK
| | - Andrew J Copp
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, London, UK
| |
Collapse
|
120
|
Kunche S, Yan H, Calof AL, Lowengrub JS, Lander AD. Feedback, Lineages and Self-Organizing Morphogenesis. PLoS Comput Biol 2016; 12:e1004814. [PMID: 26989903 PMCID: PMC4798729 DOI: 10.1371/journal.pcbi.1004814] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 02/15/2016] [Indexed: 01/31/2023] Open
Abstract
Feedback regulation of cell lineage progression plays an important role in tissue size homeostasis, but whether such feedback also plays an important role in tissue morphogenesis has yet to be explored. Here we use mathematical modeling to show that a particular feedback architecture in which both positive and negative diffusible signals act on stem and/or progenitor cells leads to the appearance of bistable or bi-modal growth behaviors, ultrasensitivity to external growth cues, local growth-driven budding, self-sustaining elongation, and the triggering of self-organization in the form of lamellar fingers. Such behaviors arise not through regulation of cell cycle speeds, but through the control of stem or progenitor self-renewal. Even though the spatial patterns that arise in this setting are the result of interactions between diffusible factors with antagonistic effects, morphogenesis is not the consequence of Turing-type instabilities.
Collapse
Affiliation(s)
- Sameeran Kunche
- Department of Biomedical Engineering, University of California, Irvine, Irvine, California, United States of America
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, United States of America
| | - Huaming Yan
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, United States of America
- Department of Mathematics, University of California, Irvine, Irvine, California, United States of America
| | - Anne L. Calof
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, United States of America
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, United States of America
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, California, United States of America
- * E-mail: (ALC); (JSL); (ADL)
| | - John S. Lowengrub
- Department of Biomedical Engineering, University of California, Irvine, Irvine, California, United States of America
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, United States of America
- Department of Mathematics, University of California, Irvine, Irvine, California, United States of America
- * E-mail: (ALC); (JSL); (ADL)
| | - Arthur D. Lander
- Department of Biomedical Engineering, University of California, Irvine, Irvine, California, United States of America
- Center for Complex Biological Systems, University of California, Irvine, Irvine, California, United States of America
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, United States of America
- * E-mail: (ALC); (JSL); (ADL)
| |
Collapse
|
121
|
Dailey SC, Febrero Planas R, Rossell Espier A, Garcia-Fernàndez J, Somorjai IML. Asymmetric Distribution of pl10 and bruno2, New Members of a Conserved Core of Early Germline Determinants in Cephalochordates. Front Ecol Evol 2016. [DOI: 10.3389/fevo.2015.00156] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
|
122
|
Kimelman D. Tales of Tails (and Trunks): Forming the Posterior Body in Vertebrate Embryos. Curr Top Dev Biol 2016; 116:517-36. [PMID: 26970638 DOI: 10.1016/bs.ctdb.2015.12.008] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A major question in developmental biology is how the early embryonic axes are established. Recent studies using different model organisms and mammalian in vitro systems have revealed the surprising result that most of the early posterior embryonic body forms from a Wnt-regulated bipotential neuromesodermal progenitor population that escapes early germ layer patterning. Part of the regulatory network that drives the maintenance and differentiation of these progenitors has recently been determined, but much remains to be discovered. This review discusses some of the common features present in all vertebrates, as well as unique aspects that different species utilize to establish their anterior-posterior (A-P) axis.
Collapse
Affiliation(s)
- David Kimelman
- Department of Biochemistry, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
123
|
Wymeersch FJ, Huang Y, Blin G, Cambray N, Wilkie R, Wong FCK, Wilson V. Position-dependent plasticity of distinct progenitor types in the primitive streak. eLife 2016; 5:e10042. [PMID: 26780186 PMCID: PMC4798969 DOI: 10.7554/elife.10042] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 01/14/2016] [Indexed: 12/22/2022] Open
Abstract
The rostrocaudal (head-to-tail) axis is supplied by populations of progenitors at the caudal end of the embryo. Despite recent advances characterising one of these populations, the neuromesodermal progenitors, their nature and relationship to other populations remains unclear. Here we show that neuromesodermal progenitors are a single Sox2lowTlow entity whose choice of neural or mesodermal fate is dictated by their position in the progenitor region. The choice of mesoderm fate is Wnt/β-catenin dependent. Wnt/β-catenin signalling is also required for a previously unrecognised phase of progenitor expansion during mid-trunk formation. Lateral/ventral mesoderm progenitors represent a distinct committed state that is unable to differentiate to neural fates, even upon overexpression of the neural transcription factor Sox2. They do not require Wnt/β-catenin signalling for mesoderm differentiation. This information aids the correct interpretation of in vivo genetic studies and the development of in vitro protocols for generating physiologically-relevant cell populations of clinical interest. DOI:http://dx.doi.org/10.7554/eLife.10042.001 Our bodies, like those of all animals with a backbone, form during embryo development in a head-to-tail sequence. This process is fuelled by populations of proliferating cells called progenitor cells, which are found in an early embryonic structure called the primitive streak, and later at the tail-end of the embryo. One of these populations – known as the neuromesodermal progenitors (or NMPs) – produces the animal’s spinal cord, muscle and bone tissue. However, it is not clear how this cell population is maintained or what triggers these cells to specialise into the correct cell type. It is even unclear whether NMPs are a single cell type or a collection of several types of progenitor, each with a slightly different propensity to make spinal cord or muscle and bone. Answering these questions could inform the future development of cell-replacement therapies for conditions such as spinal injuries. Wymeersch et al. used a range of techniques to identify, map the fate, and assess the developmental potential of progenitors in the primitive streak. This revealed fine-grained differences in the fates adopted by cells in the progenitor region. However, these regional differences were found to result from the progenitor cells’ extensive ability to respond to signals they receive from their environment, rather than being hard-wired into the progenitor cells. In fact, Wymeersch et al. detected only two distinct cell types: the NMPs and a new cell population termed lateral/paraxial mesoderm progenitors (or LPMPs), which, unlike NMPs, do not form nerve cells. Further experiments investigated the molecular signals present in the environment of these progenitors that help to decide their fate. NMPs respond to an important developmental signal, called Wnt, by adopting a so-called mesoderm fate. This signal also induces NMPs to undergo a previously unknown phase of proliferation during the formation of the animal’s body. LPMPs, on the other hand, do not require Wnt to form mesoderm. These findings show that studies with embryos can identify new progenitor populations that might be clinically relevant, and reveal new ways in which a cell’s environment inside an embryo can determine its fate. DOI:http://dx.doi.org/10.7554/eLife.10042.002
Collapse
Affiliation(s)
- Filip J Wymeersch
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Yali Huang
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Guillaume Blin
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Noemí Cambray
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Ron Wilkie
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Frederick C K Wong
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Valerie Wilson
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
124
|
Martin BL. Factors that coordinate mesoderm specification from neuromesodermal progenitors with segmentation during vertebrate axial extension. Semin Cell Dev Biol 2016; 49:59-67. [DOI: 10.1016/j.semcdb.2015.11.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 11/25/2015] [Accepted: 11/26/2015] [Indexed: 12/15/2022]
|
125
|
Mallo M. Revisiting the involvement of signaling gradients in somitogenesis. FEBS J 2015; 283:1430-7. [DOI: 10.1111/febs.13622] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 11/19/2015] [Accepted: 12/03/2015] [Indexed: 12/24/2022]
Affiliation(s)
- Moisés Mallo
- Instituto Gulbenkian de Ciencia; Oeiras Portugal
| |
Collapse
|
126
|
Row RH, Tsotras SR, Goto H, Martin BL. The zebrafish tailbud contains two independent populations of midline progenitor cells that maintain long-term germ layer plasticity and differentiate in response to local signaling cues. Development 2015; 143:244-54. [PMID: 26674311 DOI: 10.1242/dev.129015] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 12/09/2015] [Indexed: 12/25/2022]
Abstract
Vertebrate body axis formation depends on a population of bipotential neuromesodermal cells along the posterior wall of the tailbud that make a germ layer decision after gastrulation to form spinal cord and mesoderm. Despite exhibiting germ layer plasticity, these cells never give rise to midline tissues of the notochord, floor plate and dorsal endoderm, raising the question of whether midline tissues also arise from basal posterior progenitors after gastrulation. We show in zebrafish that local posterior signals specify germ layer fate in two basal tailbud midline progenitor populations. Wnt signaling induces notochord within a population of notochord/floor plate bipotential cells through negative transcriptional regulation of sox2. Notch signaling, required for hypochord induction during gastrulation, continues to act in the tailbud to specify hypochord from a notochord/hypochord bipotential cell population. Our results lend strong support to a continuous allocation model of midline tissue formation in zebrafish, and provide an embryological basis for zebrafish and mouse bifurcated notochord phenotypes as well as the rare human congenital split notochord syndrome. We demonstrate developmental equivalency between the tailbud progenitor cell populations. Midline progenitors can be transfated from notochord to somite fate after gastrulation by ectopic expression of msgn1, a master regulator of paraxial mesoderm fate, or if transplanted into the bipotential progenitors that normally give rise to somites. Our results indicate that the entire non-epidermal posterior body is derived from discrete, basal tailbud cell populations. These cells remain receptive to extracellular cues after gastrulation and continue to make basic germ layer decisions.
Collapse
Affiliation(s)
- Richard H Row
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | - Steve R Tsotras
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | - Hana Goto
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| | - Benjamin L Martin
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794-5215, USA
| |
Collapse
|
127
|
Vincent CD, Rost F, Masselink W, Brusch L, Tanaka EM. Cellular dynamics underlying regeneration of appropriate segment number during axolotl tail regeneration. BMC DEVELOPMENTAL BIOLOGY 2015; 15:48. [PMID: 26647066 PMCID: PMC4673748 DOI: 10.1186/s12861-015-0098-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 12/01/2015] [Indexed: 01/08/2023]
Abstract
Background Salamanders regenerate their tails after amputation anywhere along their length. How the system faithfully reconstitutes the original number of segments and length is not yet known. Methods To gain quantitative insight into how the system regenerates the appropriate length, we amputated tails at 4 or 16 myotomes post-cloaca and measured blastema size, cell cycle kinetics via cumulative Bromodeoxyuridine (BrdU) incorporation and the method of Nowakowski, and myotome differentiation rate. Results In early stages until day 15, blastema cells were all proliferative and divided at the same rate at both amputation levels. A larger blastema was formed in 4th versus 16th myotome amputations indicating a larger founding population. Myotome differentiation started at the same timepoint in the 4th and 16 th level blastemas. The rate of myotome formation was more rapid in 4th myotome blastemas so that by day 21 the residual blastema from the two amputation levels achieved equivalent size. At that time point, only a fraction of blastema cells remain in cycle, with the 4th myotome blastema harboring double the number of cycling cells as the 16th myotome blastema allowing it to grow faster and further reconstitute the larger number of missing myotomes. Conclusions These data suggest that there are two separable phases of blastema growth. The first is level-independent, with cells displaying unrestrained proliferation. In the second phase, the level-specific growth is revealed, where differing fractions of cells remain in the cell cycle over time.
Collapse
Affiliation(s)
- Carr D Vincent
- Max-Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 105, 01307, Dresden, Germany. .,Technische Universität Dresden, DFG Center for Regenerative Therapies, Fetscherstraße 105, 01307, Dresden, Germany.
| | - Fabian Rost
- Technische Universität Dresden, Center for Information Services and High Performance Computing, and Center for Advancing Electronics Dresden (cfaed), 01062, Dresden, Germany
| | - Wouter Masselink
- Technische Universität Dresden, DFG Center for Regenerative Therapies, Fetscherstraße 105, 01307, Dresden, Germany
| | - Lutz Brusch
- Technische Universität Dresden, Center for Information Services and High Performance Computing, and Center for Advancing Electronics Dresden (cfaed), 01062, Dresden, Germany
| | - Elly M Tanaka
- Technische Universität Dresden, DFG Center for Regenerative Therapies, Fetscherstraße 105, 01307, Dresden, Germany.
| |
Collapse
|
128
|
Henrique D, Abranches E, Verrier L, Storey KG. Neuromesodermal progenitors and the making of the spinal cord. Development 2015; 142:2864-75. [PMID: 26329597 PMCID: PMC4958456 DOI: 10.1242/dev.119768] [Citation(s) in RCA: 226] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Neuromesodermal progenitors (NMps) contribute to both the elongating spinal cord and the adjacent paraxial mesoderm. It has been assumed that these cells arise as a result of patterning of the anterior neural plate. However, as the molecular mechanisms that specify NMps in vivo are uncovered, and as protocols for generating these bipotent cells from mouse and human pluripotent stem cells in vitro are established, the emerging data suggest that this view needs to be revised. Here, we review the characteristics, regulation, in vitro derivation and in vivo induction of NMps. We propose that these cells arise within primitive streak-associated epiblast via a mechanism that is separable from that which establishes neural fate in the anterior epiblast. We thus argue for the existence of two distinct routes for making central nervous system progenitors.
Collapse
Affiliation(s)
- Domingos Henrique
- Instituto de Medicina Molecular and Instituto de Histologia e Biologia do Desenvolvimento, Faculdade de Medicina da Universidade de Lisboa, Avenida Prof. Egas Moniz, Lisboa 1649-028, Portugal
| | - Elsa Abranches
- Instituto de Medicina Molecular and Instituto de Histologia e Biologia do Desenvolvimento, Faculdade de Medicina da Universidade de Lisboa, Avenida Prof. Egas Moniz, Lisboa 1649-028, Portugal
| | - Laure Verrier
- Division of Cell & Developmental Biology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Kate G Storey
- Division of Cell & Developmental Biology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| |
Collapse
|
129
|
Pirmoazen E, Matin M, Najafzadeh N, Golmohammadi MG, Sagha M. Retinoic acid recapitulates the action of the somites on neural differentiation of the developing caudal neural plate in chick embryo. NEUROCHEM J+ 2015. [DOI: 10.1134/s1819712415040133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
130
|
Rodrigo Albors A, Tazaki A, Rost F, Nowoshilow S, Chara O, Tanaka EM. Planar cell polarity-mediated induction of neural stem cell expansion during axolotl spinal cord regeneration. eLife 2015; 4:e10230. [PMID: 26568310 PMCID: PMC4755742 DOI: 10.7554/elife.10230] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 11/12/2015] [Indexed: 02/07/2023] Open
Abstract
Axolotls are uniquely able to mobilize neural stem cells to regenerate all missing regions of the spinal cord. How a neural stem cell under homeostasis converts after injury to a highly regenerative cell remains unknown. Here, we show that during regeneration, axolotl neural stem cells repress neurogenic genes and reactivate a transcriptional program similar to embryonic neuroepithelial cells. This dedifferentiation includes the acquisition of rapid cell cycles, the switch from neurogenic to proliferative divisions, and the re-expression of planar cell polarity (PCP) pathway components. We show that PCP induction is essential to reorient mitotic spindles along the anterior-posterior axis of elongation, and orthogonal to the cell apical-basal axis. Disruption of this property results in premature neurogenesis and halts regeneration. Our findings reveal a key role for PCP in coordinating the morphogenesis of spinal cord outgrowth with the switch from a homeostatic to a regenerative stem cell that restores missing tissue. DOI:http://dx.doi.org/10.7554/eLife.10230.001 Stem cells found in adult tissues are vitally important for tissue repair and maintenance. These cells divide in two main ways: equally to create two new stem cells, or unequally to create a stem cell and a cell that can develop into one of the cell types in the tissue. A key challenge for biologists is to understand how these tissue-resident stem cells are activated and organized to regenerate injured or missing tissue. Throughout the life of the axolotl salamander, neural stem cells in the spinal cord occasionally divide to add new nerve cells to the healthy spinal cord. However, the axolotl can also regenerate part of its spinal cord, for example if its tail is lost. Under these conditions, the neural stem cells can convert into a highly regenerative stem cell that can produce all the different cell types needed to regrow the spinal cord. As a stem cell becomes a new cell type, it activates different sets of genes. Therefore, Rodrigo Albors, Tazaki et al. measured gene activity in the neural stem cells involved in axolotl spinal cord regeneration to uncover how these cells develop into a more regenerative form. This revealed that when an axolotl tail is amputated, resident stem cells turn off the genes that are specifically active in neuron-generating cells. In addition, they activate a similar set of genes to that seen in the embryonic cells that form the developing nervous system. These genes speed up cell division and activate an important signaling pathway. This pathway – the Wnt/PCP pathway – fulfils various developmental roles, one being to orient cell divisions, particularly in elongating tissues. In axolotls, this pathway causes the stem cells to divide equally to increase the number of available stem cells, and orients the direction of these divisions to ensure that the regenerating spinal cord elongates correctly. If this pathway is disrupted, the cells return to dividing unequally, generating nerve cells prematurely and halting the growth of the spinal cord. Such insights could help develop methods of repairing damaged nervous tissue in other animals that cannot regenerate to the extent that axolotls can. DOI:http://dx.doi.org/10.7554/eLife.10230.002
Collapse
Affiliation(s)
- Aida Rodrigo Albors
- Deutsche Forschungsgemeinschaft - Center for Regenerative Therapies Dresden, Dresden, Germany.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Technische Universität Dresden, Dresden, Germany
| | - Akira Tazaki
- Deutsche Forschungsgemeinschaft - Center for Regenerative Therapies Dresden, Dresden, Germany.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Technische Universität Dresden, Dresden, Germany
| | - Fabian Rost
- Center for Information Services and High Performance Computing, Technische Universität Dresden, Dresden, Germany
| | - Sergej Nowoshilow
- Deutsche Forschungsgemeinschaft - Center for Regenerative Therapies Dresden, Dresden, Germany.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Technische Universität Dresden, Dresden, Germany
| | - Osvaldo Chara
- Center for Information Services and High Performance Computing, Technische Universität Dresden, Dresden, Germany.,Institute of Physics of Liquids and Biological Systems, National Scientific and Technical Research Council, University of La Plata, La Plata, Argentina
| | - Elly M Tanaka
- Deutsche Forschungsgemeinschaft - Center for Regenerative Therapies Dresden, Dresden, Germany.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
131
|
Bertrand S, Aldea D, Oulion S, Subirana L, de Lera AR, Somorjai I, Escriva H. Evolution of the Role of RA and FGF Signals in the Control of Somitogenesis in Chordates. PLoS One 2015; 10:e0136587. [PMID: 26371756 PMCID: PMC4570818 DOI: 10.1371/journal.pone.0136587] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 08/05/2015] [Indexed: 11/18/2022] Open
Abstract
During vertebrate development, the paraxial mesoderm becomes segmented, forming somites that will give rise to dermis, axial skeleton and skeletal muscles. Although recently challenged, the "clock and wavefront" model for somitogenesis explains how interactions between several cell-cell communication pathways, including the FGF, RA, Wnt and Notch signals, control the formation of these bilateral symmetric blocks. In the cephalochordate amphioxus, which belongs to the chordate phylum together with tunicates and vertebrates, the dorsal paraxial mesendoderm also periodically forms somites, although this process is asymmetric and extends along the whole body. It has been previously shown that the formation of the most anterior somites in amphioxus is dependent upon FGF signalling. However, the signals controlling somitogenesis during posterior elongation in amphioxus are still unknown. Here we show that, contrary to vertebrates, RA and FGF signals act independently during posterior elongation and that they are not mandatory for posterior somites to form. Moreover, we show that RA is not able to buffer the left/right asymmetry machinery that is controlled through the asymmetric expression of Nodal pathway actors. Our results give new insights into the evolution of the somitogenesis process in chordates. They suggest that RA and FGF pathways have acquired specific functions in the control of somitogenesis in vertebrates. We propose that the "clock and wavefront" system was selected specifically in vertebrates in parallel to the development of more complex somite-derived structures but that it was not required for somitogenesis in the ancestor of chordates.
Collapse
Affiliation(s)
- Stéphanie Bertrand
- UPMC Univ Paris 06, UMR 7232, BIOM, Observatoire Océanologique de Banyuls sur Mer, F-66650, Banyuls/Mer, France
- * E-mail: (SB); (HE)
| | - Daniel Aldea
- UPMC Univ Paris 06, UMR 7232, BIOM, Observatoire Océanologique de Banyuls sur Mer, F-66650, Banyuls/Mer, France
| | - Silvan Oulion
- UPMC Univ Paris 06, UMR 7232, BIOM, Observatoire Océanologique de Banyuls sur Mer, F-66650, Banyuls/Mer, France
| | - Lucie Subirana
- UPMC Univ Paris 06, UMR 7232, BIOM, Observatoire Océanologique de Banyuls sur Mer, F-66650, Banyuls/Mer, France
| | - Angel R. de Lera
- Departamento de Química Orgánica, Facultade de Química, CINBIO, Universidade de Vigo, and Instituto de Investigación Biomédica de Vigo (IBIV), Vigo, Spain
| | - Ildiko Somorjai
- UPMC Univ Paris 06, UMR 7232, BIOM, Observatoire Océanologique de Banyuls sur Mer, F-66650, Banyuls/Mer, France
| | - Hector Escriva
- UPMC Univ Paris 06, UMR 7232, BIOM, Observatoire Océanologique de Banyuls sur Mer, F-66650, Banyuls/Mer, France
- * E-mail: (SB); (HE)
| |
Collapse
|
132
|
Cunningham TJ, Brade T, Sandell LL, Lewandoski M, Trainor PA, Colas A, Mercola M, Duester G. Retinoic Acid Activity in Undifferentiated Neural Progenitors Is Sufficient to Fulfill Its Role in Restricting Fgf8 Expression for Somitogenesis. PLoS One 2015; 10:e0137894. [PMID: 26368825 PMCID: PMC4569375 DOI: 10.1371/journal.pone.0137894] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 08/24/2015] [Indexed: 12/20/2022] Open
Abstract
Bipotent axial stem cells residing in the caudal epiblast during late gastrulation generate neuroectodermal and presomitic mesodermal progeny that coordinate somitogenesis with neural tube formation, but the mechanism that controls these two fates is not fully understood. Retinoic acid (RA) restricts the anterior extent of caudal fibroblast growth factor 8 (Fgf8) expression in both mesoderm and neural plate to control somitogenesis and neurogenesis, however it remains unclear where RA acts to control the spatial expression of caudal Fgf8. Here, we found that mouse Raldh2-/- embryos, lacking RA synthesis and displaying a consistent small somite defect, exhibited abnormal expression of key markers of axial stem cell progeny, with decreased Sox2+ and Sox1+ neuroectodermal progeny and increased Tbx6+ presomitic mesodermal progeny. The Raldh2-/- small somite defect was rescued by treatment with an FGF receptor antagonist. Rdh10 mutants, with a less severe RA synthesis defect, were found to exhibit a small somite defect and anterior expansion of caudal Fgf8 expression only for somites 1-6, with normal somite size and Fgf8 expression thereafter. Rdh10 mutants were found to lack RA activity during the early phase when somites are small, but at the 6-somite stage RA activity was detected in neural plate although not in presomitic mesoderm. Expression of a dominant-negative RA receptor in mesoderm eliminated RA activity in presomitic mesoderm but did not affect somitogenesis. Thus, RA activity in the neural plate is sufficient to prevent anterior expansion of caudal Fgf8 expression associated with a small somite defect. Our studies provide evidence that RA restriction of Fgf8 expression in undifferentiated neural progenitors stimulates neurogenesis while also restricting the anterior extent of the mesodermal Fgf8 mRNA gradient that controls somite size, providing new insight into the mechanism that coordinates somitogenesis with neurogenesis.
Collapse
Affiliation(s)
- Thomas J. Cunningham
- Development, Aging, and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Thomas Brade
- Development, Aging, and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Lisa L. Sandell
- Department of Molecular, Cellular, and Craniofacial Biology, University of Louisville, Louisville, Kentucky, United States of America
| | - Mark Lewandoski
- Laboratory of Cancer and Developmental Biology, National Cancer Institute, National Institutes of Health, Frederick, Maryland, United States of America
| | - Paul A. Trainor
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Alexandre Colas
- Development, Aging, and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
- Department of Bioengineering, University of California at San Diego, La Jolla, California, United States of America
| | - Mark Mercola
- Development, Aging, and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
- Department of Bioengineering, University of California at San Diego, La Jolla, California, United States of America
| | - Gregg Duester
- Development, Aging, and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
133
|
Garriock RJ, Chalamalasetty RB, Kennedy MW, Canizales LC, Lewandoski M, Yamaguchi TP. Lineage tracing of neuromesodermal progenitors reveals novel Wnt-dependent roles in trunk progenitor cell maintenance and differentiation. Development 2015; 142:1628-38. [PMID: 25922526 DOI: 10.1242/dev.111922] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In the development of the vertebrate body plan, Wnt3a is thought to promote the formation of paraxial mesodermal progenitors (PMPs) of the trunk region while suppressing neural specification. Recent lineage-tracing experiments have demonstrated that these trunk neural progenitors and PMPs derive from a common multipotent progenitor called the neuromesodermal progenitor (NMP). NMPs are known to reside in the anterior primitive streak (PS) region; however, the extent to which NMPs populate the PS and contribute to the vertebrate body plan, and the precise role that Wnt3a plays in regulating NMP self-renewal and differentiation are unclear. To address this, we used cell-specific markers (Sox2 and T) and tamoxifen-induced Cre recombinase-based lineage tracing to locate putative NMPs in vivo. We provide functional evidence for NMP location primarily in the epithelial PS, and to a lesser degree in the ingressed PS. Lineage-tracing studies in Wnt3a/β-catenin signaling pathway mutants provide genetic evidence that trunk progenitors normally fated to enter the mesodermal germ layer can be redirected towards the neural lineage. These data, combined with previous PS lineage-tracing studies, support a model that epithelial anterior PS cells are Sox2(+)T(+) multipotent NMPs and form the bulk of neural progenitors and PMPs of the posterior trunk region. Finally, we find that Wnt3a/β-catenin signaling directs trunk progenitors towards PMP fates; however, our data also suggest that Wnt3a positively supports a progenitor state for both mesodermal and neural progenitors.
Collapse
Affiliation(s)
- Robert J Garriock
- Center for Cancer Research, Cancer and Developmental Biology Laboratory, Cell Signaling in Vertebrate Development Section, NCI-Frederick, NIH, Frederick, MD 21702, USA
| | - Ravindra B Chalamalasetty
- Center for Cancer Research, Cancer and Developmental Biology Laboratory, Cell Signaling in Vertebrate Development Section, NCI-Frederick, NIH, Frederick, MD 21702, USA
| | - Mark W Kennedy
- Center for Cancer Research, Cancer and Developmental Biology Laboratory, Cell Signaling in Vertebrate Development Section, NCI-Frederick, NIH, Frederick, MD 21702, USA
| | - Lauren C Canizales
- Center for Cancer Research, Cancer and Developmental Biology Laboratory, Cell Signaling in Vertebrate Development Section, NCI-Frederick, NIH, Frederick, MD 21702, USA
| | - Mark Lewandoski
- Center for Cancer Research, Cancer and Developmental Biology Laboratory, Cell Signaling in Vertebrate Development Section, NCI-Frederick, NIH, Frederick, MD 21702, USA
| | - Terry P Yamaguchi
- Center for Cancer Research, Cancer and Developmental Biology Laboratory, Cell Signaling in Vertebrate Development Section, NCI-Frederick, NIH, Frederick, MD 21702, USA
| |
Collapse
|
134
|
Faial T, Bernardo AS, Mendjan S, Diamanti E, Ortmann D, Gentsch GE, Mascetti VL, Trotter MWB, Smith JC, Pedersen RA. Brachyury and SMAD signalling collaboratively orchestrate distinct mesoderm and endoderm gene regulatory networks in differentiating human embryonic stem cells. Development 2015; 142:2121-35. [PMID: 26015544 PMCID: PMC4483767 DOI: 10.1242/dev.117838] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 04/30/2015] [Indexed: 12/17/2022]
Abstract
The transcription factor brachyury (T, BRA) is one of the first markers of gastrulation and lineage specification in vertebrates. Despite its wide use and importance in stem cell and developmental biology, its functional genomic targets in human cells are largely unknown. Here, we use differentiating human embryonic stem cells to study the role of BRA in activin A-induced endoderm and BMP4-induced mesoderm progenitors. We show that BRA has distinct genome-wide binding landscapes in these two cell populations, and that BRA interacts and collaborates with SMAD1 or SMAD2/3 signalling to regulate the expression of its target genes in a cell-specific manner. Importantly, by manipulating the levels of BRA in cells exposed to different signalling environments, we demonstrate that BRA is essential for mesoderm but not for endoderm formation. Together, our data illuminate the function of BRA in the context of human embryonic development and show that the regulatory role of BRA is context dependent. Our study reinforces the importance of analysing the functions of a transcription factor in different cellular and signalling environments.
Collapse
Affiliation(s)
- Tiago Faial
- The Anne McLaren Laboratory for Regenerative Medicine, Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0SZ, UK The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, London NW7 1AA, UK Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
| | - Andreia S Bernardo
- The Anne McLaren Laboratory for Regenerative Medicine, Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0SZ, UK The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, London NW7 1AA, UK Department of Surgery, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Sasha Mendjan
- The Anne McLaren Laboratory for Regenerative Medicine, Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0SZ, UK Department of Surgery, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Evangelia Diamanti
- Cambridge Institute for Medical Research and Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0XY, UK
| | - Daniel Ortmann
- The Anne McLaren Laboratory for Regenerative Medicine, Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0SZ, UK Department of Surgery, University of Cambridge, Cambridge CB2 0QQ, UK
| | - George E Gentsch
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, London NW7 1AA, UK Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
| | - Victoria L Mascetti
- The Anne McLaren Laboratory for Regenerative Medicine, Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0SZ, UK Department of Surgery, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Matthew W B Trotter
- The Anne McLaren Laboratory for Regenerative Medicine, Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0SZ, UK
| | - James C Smith
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, London NW7 1AA, UK Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
| | - Roger A Pedersen
- The Anne McLaren Laboratory for Regenerative Medicine, Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0SZ, UK Department of Surgery, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
135
|
Bouldin CM, Manning AJ, Peng YH, Farr GH, Hung KL, Dong A, Kimelman D. Wnt signaling and tbx16 form a bistable switch to commit bipotential progenitors to mesoderm. Development 2015; 142:2499-507. [PMID: 26062939 DOI: 10.1242/dev.124024] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 06/03/2015] [Indexed: 01/16/2023]
Abstract
Anterior to posterior growth of the vertebrate body is fueled by a posteriorly located population of bipotential neuro-mesodermal progenitor cells. These progenitors have a limited rate of proliferation and their maintenance is crucial for completion of the anterior-posterior axis. How they leave the progenitor state and commit to differentiation is largely unknown, in part because widespread modulation of factors essential for this process causes organism-wide effects. Using a novel assay, we show that zebrafish Tbx16 (Spadetail) is capable of advancing mesodermal differentiation cell-autonomously. Tbx16 locks cells into the mesodermal state by not only activating downstream mesodermal genes, but also by repressing bipotential progenitor genes, in part through a direct repression of sox2. We demonstrate that tbx16 is activated as cells move from an intermediate Wnt environment to a high Wnt environment, and show that Wnt signaling activates the tbx16 promoter. Importantly, high-level Wnt signaling is able to accelerate mesodermal differentiation cell-autonomously, just as we observe with Tbx16. Finally, because our assay for mesodermal commitment is quantitative we are able to show that the acceleration of mesodermal differentiation is surprisingly incomplete, implicating a potential separation of cell movement and differentiation during this process. Together, our data suggest a model in which high levels of Wnt signaling induce a transition to mesoderm by directly activating tbx16, which in turn acts to irreversibly flip a bistable switch, leading to maintenance of the mesodermal fate and repression of the bipotential progenitor state, even as cells leave the initial high-Wnt environment.
Collapse
Affiliation(s)
- Cortney M Bouldin
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Alyssa J Manning
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Yu-Hsuan Peng
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Gist H Farr
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - King L Hung
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Alice Dong
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - David Kimelman
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
136
|
Current Bioengineering Methods for Whole Kidney Regeneration. Stem Cells Int 2015; 2015:724047. [PMID: 26089921 PMCID: PMC4452081 DOI: 10.1155/2015/724047] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 01/13/2015] [Accepted: 01/13/2015] [Indexed: 02/07/2023] Open
Abstract
Kidney regeneration is likely to provide an inexhaustible source of tissues and organs for immunosuppression-free transplantation. It is currently garnering considerable attention and might replace kidney dialysis as the ultimate therapeutic strategy for renal failure. However, anatomical complications make kidney regeneration difficult. Here, we review recent advances in the field of kidney regeneration, including (i) the directed differentiation of induced pluripotent stem cells/embryonic stem cells into kidney cells; (ii) blastocyst decomplementation; (iii) use of a decellularized cadaveric scaffold; (iv) embryonic organ transplantation; and (v) use of a nephrogenic niche for growing xenoembryos for de novo kidney regeneration from stem cells. All these approaches represent potentially promising therapeutic strategies for the treatment of patients with chronic kidney disease. Although many obstacles to kidney regeneration remain, we hope that innovative strategies and reliable research will ultimately allow the restoration of renal function in patients with end-stage kidney disease.
Collapse
|
137
|
Abstract
Spina bifida is a birth defect in which the vertebral column is open, often with spinal cord involvement. The most clinically significant subtype is myelomeningocele (open spina bifida), which is a condition characterized by failure of the lumbosacral spinal neural tube to close during embryonic development. The exposed neural tissue degenerates in utero, resulting in neurological deficit that varies with the level of the lesion. Occurring in approximately 1 per 1,000 births worldwide, myelomeningocele is one of the most common congenital malformations, but its cause is largely unknown. The genetic component is estimated at 60-70%, but few causative genes have been identified to date, despite much information from mouse models. Non-genetic maternal risk factors include reduced folate intake, anticonvulsant therapy, diabetes mellitus and obesity. Primary prevention by periconceptional supplementation with folic acid has been demonstrated in clinical trials, leading to food fortification programmes in many countries. Prenatal diagnosis is achieved by ultrasonography, enabling women to seek termination of pregnancy. Individuals who survive to birth have their lesions closed surgically, with subsequent management of associated defects, including the Chiari II brain malformation, hydrocephalus, and urological and orthopaedic sequelae. Fetal surgical repair of myelomeningocele has been associated with improved early neurological outcome compared with postnatal operation. Myelomeningocele affects quality of life during childhood, adolescence and adulthood, posing a challenge for individuals, families and society as a whole. For an illustrated summary of this Primer, visit: http://go.nature.com/fK9XNa.
Collapse
|
138
|
Cunningham TJ, Kumar S, Yamaguchi TP, Duester G. Wnt8a and Wnt3a cooperate in the axial stem cell niche to promote mammalian body axis extension. Dev Dyn 2015; 244:797-807. [PMID: 25809880 DOI: 10.1002/dvdy.24275] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 03/13/2015] [Accepted: 03/16/2015] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Vertebrate body axis extension occurs in a head-to-tail direction from a caudal progenitor zone that responds to interacting signals. Wnt/β-catenin signaling is critical for generation of paraxial mesoderm, somite formation, and maintenance of the axial stem cell pool. Body axis extension requires Wnt8a in lower vertebrates, but in mammals Wnt3a is required, although the anterior trunk develops in the absence of Wnt3a. RESULTS We examined mouse Wnt8a(-/-) and Wnt3a(-/-) single and double mutants to explore whether mammalian Wnt8a contributes to body axis extension and to determine whether a posterior growth function for Wnt8a is conserved throughout the vertebrate lineage. We find that caudal Wnt8a is expressed only during early somite stages and is required for normal development of the anterior trunk in the absence of Wnt3a. During this time, we show that Wnt8a and Wnt3a cooperate to maintain Fgf8 expression and prevent premature Sox2 up-regulation in the axial stem cell niche, critical for posterior growth. Similar to Fgf8, Wnt8a requires retinoic acid (RA) signaling to restrict its caudal expression boundary and possesses an upstream RA response element that binds RA receptors. CONCLUSIONS These findings provide new insight into interaction of caudal Wnt-FGF-RA signals required for body axis extension.
Collapse
Affiliation(s)
- Thomas J Cunningham
- Development, Aging, and Regeneration Program, Sanford-Burnham Medical Research Institute, La Jolla, California
| | - Sandeep Kumar
- Development, Aging, and Regeneration Program, Sanford-Burnham Medical Research Institute, La Jolla, California
| | - Terry P Yamaguchi
- Cancer and Developmental Biology Laboratory, NCI-Frederick, National Institutes of Health, Frederick, Maryland
| | - Gregg Duester
- Development, Aging, and Regeneration Program, Sanford-Burnham Medical Research Institute, La Jolla, California
| |
Collapse
|
139
|
Gouti M, Metzis V, Briscoe J. The route to spinal cord cell types: a tale of signals and switches. Trends Genet 2015; 31:282-9. [PMID: 25823696 DOI: 10.1016/j.tig.2015.03.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 02/28/2015] [Accepted: 03/02/2015] [Indexed: 01/20/2023]
Abstract
Understanding the mechanisms that control induction and elaboration of the vertebrate central nervous system (CNS) requires an analysis of the extrinsic signals and downstream transcriptional networks that assign cell fates in the correct space and time. We focus on the generation and patterning of the spinal cord. We summarize evidence that the origin of the spinal cord is distinct from the anterior regions of the CNS. We discuss how this affects the gene regulatory networks and cell state transitions that specify spinal cord cell subtypes, and we highlight how the timing of extracellular signals and dynamic control of transcriptional networks contribute to the correct spatiotemporal generation of different neural cell types.
Collapse
Affiliation(s)
- Mina Gouti
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, NW7 1AA, UK
| | - Vicki Metzis
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, NW7 1AA, UK
| | - James Briscoe
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, Mill Hill, London, NW7 1AA, UK.
| |
Collapse
|
140
|
Andre P, Song H, Kim W, Kispert A, Yang Y. Wnt5a and Wnt11 regulate mammalian anterior-posterior axis elongation. Development 2015; 142:1516-27. [PMID: 25813538 DOI: 10.1242/dev.119065] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 03/04/2015] [Indexed: 01/01/2023]
Abstract
Mesoderm formation and subsequent anterior-posterior (A-P) axis elongation are fundamental aspects of gastrulation, which is initiated by formation of the primitive streak (PS). Convergent extension (CE) movements and epithelial-mesenchymal transition (EMT) are important for A-P axis elongation in vertebrate embryos. The evolutionarily conserved planar cell polarity (PCP) pathway regulates CE, and Wnts regulate many aspects of gastrulation including CE and EMT. However, the Wnt ligands that regulate A-P axis elongation in mammalian development remain unknown. Wnt11 and Wnt5a regulate axis elongation in lower vertebrates, but only Wnt5a, not Wnt11, regulates mammalian PCP signaling and A-P axis elongation in development. Here, by generating Wnt5a; Wnt11 compound mutants, we show that Wnt11 and Wnt5a play redundant roles during mouse A-P axis elongation. Both genes regulate trunk notochord extension through PCP-controlled CE of notochord cells, establishing a role for Wnt11 in mammalian PCP. We show that Wnt5a and Wnt11 are required for proper patterning of the neural tube and somites by regulating notochord formation, and provide evidence that both genes are required for the generation and migration of axial and paraxial mesodermal precursor cells by regulating EMT. Axial and paraxial mesodermal precursors ectopically accumulate in the PS at late gastrula stages in Wnt5a(-/-); Wnt11(-/-) embryos and these cells ectopically express epithelial cell adhesion molecules. Our data suggest that Wnt5a and Wnt11 regulate EMT by inducing p38 (Mapk14) phosphorylation. Our findings provide new insights into the role of Wnt5a and Wnt11 in mouse early development and also in cancer metastasis, during which EMT plays a crucial role.
Collapse
Affiliation(s)
- Philipp Andre
- Genetic Disease Research Branch, National Human Genome Research Institute, Bethesda, MD 20814, USA
| | - Hai Song
- Genetic Disease Research Branch, National Human Genome Research Institute, Bethesda, MD 20814, USA
| | - Wantae Kim
- Genetic Disease Research Branch, National Human Genome Research Institute, Bethesda, MD 20814, USA
| | - Andreas Kispert
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover D-30625, Germany
| | - Yingzi Yang
- Genetic Disease Research Branch, National Human Genome Research Institute, Bethesda, MD 20814, USA Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Ave., Boston, MA 02115, USA
| |
Collapse
|
141
|
Denans N, Iimura T, Pourquié O. Hox genes control vertebrate body elongation by collinear Wnt repression. eLife 2015; 4. [PMID: 25719209 PMCID: PMC4384752 DOI: 10.7554/elife.04379] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 02/20/2015] [Indexed: 12/23/2022] Open
Abstract
In vertebrates, the total number of vertebrae is precisely defined. Vertebrae derive
from embryonic somites that are continuously produced posteriorly from the presomitic
mesoderm (PSM) during body formation. We show that in the chicken embryo, activation
of posterior Hox genes (paralogs 9–13) in the tail-bud
correlates with the slowing down of axis elongation. Our data indicate that a subset
of progressively more posterior Hox genes, which are collinearly
activated in vertebral precursors, repress Wnt activity with increasing strength.
This leads to a graded repression of the Brachyury/T transcription
factor, reducing mesoderm ingression and slowing down the elongation process. Due to
the continuation of somite formation, this mechanism leads to the progressive
reduction of PSM size. This ultimately brings the retinoic acid (RA)-producing
segmented region in close vicinity to the tail bud, potentially accounting for the
termination of segmentation and axis elongation. DOI:http://dx.doi.org/10.7554/eLife.04379.001 In humans and other vertebrates, the number of bones (vertebrae) in the spine is
determined early in development. The vertebrae form from blocks of tissue called
somites that make segments along the body axis—a virtual line running from the
head to the tail-end—of the embryo. The somites form as the embryo increases
in length, with new somites forming periodically at the back near the embryo's
tail-end. A family of genes called the Hox genes are involved in controlling
the formation of the somites. However, it is not known whether they directly control
the number of somites that form, or whether they control the length of the body of
the embryo. Denans et al. studied the Hox genes in chicken embryos. The
experiments suggest that the activation of some of the Hox genes in
a structure called the tail-bud, which is found at the tail-end of the embryo, slow
down the elongation of the body. The Hox genes achieve this by
repressing the activity of a signaling pathway called Wnt so that Wnt activity in the
tail-bud progressively decreases as the embryo develops. The elongation of the body stops when the levels of a molecule called retinoic acid
increase in the tail-bud, which causes the loss of the stem cells that are needed to
make the somites. Denans et al.'s findings suggest that Hox
genes influence the timing of the halt in elongation, which in turn is important for
determining the total number of somites that form. Understanding how
Hox genes control the formation of the cells that will make up
the somites and influence Wnt signaling is a major challenge for the future. DOI:http://dx.doi.org/10.7554/eLife.04379.002
Collapse
Affiliation(s)
- Nicolas Denans
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, University of Strasbourg, Illkirch, France
| | - Tadahiro Iimura
- Stowers Institute for Medical Research, Kansas City, United States
| | - Olivier Pourquié
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, University of Strasbourg, Illkirch, France
| |
Collapse
|
142
|
Cunningham TJ, Duester G. Mechanisms of retinoic acid signalling and its roles in organ and limb development. Nat Rev Mol Cell Biol 2015; 16:110-23. [PMID: 25560970 PMCID: PMC4636111 DOI: 10.1038/nrm3932] [Citation(s) in RCA: 413] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Retinoic acid (RA) signalling has a central role during vertebrate development. RA synthesized in specific locations regulates transcription by interacting with nuclear RA receptors (RARs) bound to RA response elements (RAREs) near target genes. RA was first implicated in signalling on the basis of its teratogenic effects on limb development. Genetic studies later revealed that endogenous RA promotes forelimb initiation by repressing fibroblast growth factor 8 (Fgf8). Insights into RA function in the limb serve as a paradigm for understanding how RA regulates other developmental processes. In vivo studies have identified RAREs that control repression of Fgf8 during body axis extension or activation of homeobox (Hox) genes and other key regulators during neuronal differentiation and organogenesis.
Collapse
Affiliation(s)
- Thomas J Cunningham
- Development, Aging, and Regeneration Program, Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Road, La Jolla, California 92037, USA
| | - Gregg Duester
- Development, Aging, and Regeneration Program, Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Road, La Jolla, California 92037, USA
| |
Collapse
|
143
|
Lee K, Skromne I. Retinoic acid regulates size, pattern and alignment of tissues at the head-trunk transition. Development 2015; 141:4375-84. [PMID: 25371368 DOI: 10.1242/dev.109603] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
At the head-trunk transition, hindbrain and spinal cord alignment to occipital and vertebral bones is crucial for coherent neural and skeletal system organization. Changes in neural or mesodermal tissue configuration arising from defects in the specification, patterning or relative axial placement of territories can severely compromise their integration and function. Here, we show that coordination of neural and mesodermal tissue at the zebrafish head-trunk transition crucially depends on two novel activities of the signaling factor retinoic acid (RA): one specifying the size and the other specifying the axial position relative to mesodermal structures of the hindbrain territory. These activities are each independent but coordinated with the well-established function of RA in hindbrain patterning. Using neural and mesodermal landmarks we demonstrate that the functions of RA in aligning neural and mesodermal tissues temporally precede the specification of hindbrain and spinal cord territories and the activation of hox transcription. Using cell transplantation assays we show that RA activity in the neuroepithelium regulates hindbrain patterning directly and territory size specification indirectly. This indirect function is partially dependent on Wnts but independent of FGFs. Importantly, RA specifies and patterns the hindbrain territory by antagonizing the activity of the spinal cord specification gene cdx4; loss of Cdx4 rescues the defects associated with the loss of RA, including the reduction in hindbrain size and the loss of posterior rhombomeres. We propose that at the head-trunk transition, RA coordinates specification, patterning and alignment of neural and mesodermal tissues that are essential for the organization and function of the neural and skeletal systems.
Collapse
Affiliation(s)
- Keun Lee
- Department of Biology, University of Miami, Coral Gables, FL 33146, USA
| | - Isaac Skromne
- Department of Biology, University of Miami, Coral Gables, FL 33146, USA
| |
Collapse
|
144
|
van den Brink SC, Baillie-Johnson P, Balayo T, Hadjantonakis AK, Nowotschin S, Turner DA, Martinez Arias A. Symmetry breaking, germ layer specification and axial organisation in aggregates of mouse embryonic stem cells. Development 2015; 141:4231-42. [PMID: 25371360 PMCID: PMC4302915 DOI: 10.1242/dev.113001] [Citation(s) in RCA: 296] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mouse embryonic stem cells (mESCs) are clonal populations derived from preimplantation mouse embryos that can be propagated in vitro and, when placed into blastocysts, contribute to all tissues of the embryo and integrate into the normal morphogenetic processes, i.e. they are pluripotent. However, although they can be steered to differentiate in vitro into all cell types of the organism, they cannot organise themselves into structures that resemble embryos. When aggregated into embryoid bodies they develop disorganised masses of different cell types with little spatial coherence. An exception to this rule is the emergence of retinas and anterior cortex-like structures under minimal culture conditions. These structures emerge from the cultures without any axial organisation. Here, we report that small aggregates of mESCs, of about 300 cells, self-organise into polarised structures that exhibit collective behaviours reminiscent of those that cells exhibit in early mouse embryos, including symmetry breaking, axial organisation, germ layer specification and cell behaviour, as well as axis elongation. The responses are signal specific and uncouple processes that in the embryo are tightly associated, such as specification of the anteroposterior axis and anterior neural development, or endoderm specification and axial elongation. We discuss the meaning and implications of these observations and the potential uses of these structures which, because of their behaviour, we suggest to call ‘gastruloids’.
Collapse
Affiliation(s)
| | | | - Tina Balayo
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | | | - Sonja Nowotschin
- Developmental Biology Program, Sloan-Kettering Institute, New York, NY 10065, USA
| | - David A Turner
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | | |
Collapse
|
145
|
Chalamalasetty RB, Garriock RJ, Dunty WC, Kennedy MW, Jailwala P, Si H, Yamaguchi TP. Mesogenin 1 is a master regulator of paraxial presomitic mesoderm differentiation. Development 2015; 141:4285-97. [PMID: 25371364 DOI: 10.1242/dev.110908] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Neuromesodermal (NM) stem cells generate neural and paraxial presomitic mesoderm (PSM) cells, which are the respective progenitors of the spinal cord and musculoskeleton of the trunk and tail. The Wnt-regulated basic helix-loop-helix (bHLH) transcription factor mesogenin 1 (Msgn1) has been implicated as a cooperative regulator working in concert with T-box genes to control PSM formation in zebrafish, although the mechanism is unknown. We show here that, in mice, Msgn1 alone controls PSM differentiation by directly activating the transcriptional programs that define PSM identity, epithelial-mesenchymal transition, motility and segmentation. Forced expression of Msgn1 in NM stem cells in vivo reduced the contribution of their progeny to the neural tube, and dramatically expanded the unsegmented mesenchymal PSM while blocking somitogenesis and notochord differentiation. Expression of Msgn1 was sufficient to partially rescue PSM differentiation in Wnt3a(-/-) embryos, demonstrating that Msgn1 functions downstream of Wnt3a as the master regulator of PSM differentiation. Our data provide new insights into how cell fate decisions are imposed by the expression of a single transcriptional regulator.
Collapse
Affiliation(s)
- Ravindra B Chalamalasetty
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, NCI-Frederick, NIH, Frederick, MD 21702, USA
| | - Robert J Garriock
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, NCI-Frederick, NIH, Frederick, MD 21702, USA
| | - William C Dunty
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, NCI-Frederick, NIH, Frederick, MD 21702, USA
| | - Mark W Kennedy
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, NCI-Frederick, NIH, Frederick, MD 21702, USA
| | - Parthav Jailwala
- CCRIFX Bioinformatics Core, Leidos Biomedical Research, FNLCR, Frederick, MD 21702, USA
| | - Han Si
- CCRIFX Bioinformatics Core, Leidos Biomedical Research, FNLCR, Frederick, MD 21702, USA
| | - Terry P Yamaguchi
- Cancer and Developmental Biology Laboratory, Center for Cancer Research, NCI-Frederick, NIH, Frederick, MD 21702, USA
| |
Collapse
|
146
|
Turner DA, Hayward PC, Baillie-Johnson P, Rué P, Broome R, Faunes F, Martinez Arias A. Wnt/β-catenin and FGF signalling direct the specification and maintenance of a neuromesodermal axial progenitor in ensembles of mouse embryonic stem cells. Development 2015; 141:4243-53. [PMID: 25371361 PMCID: PMC4302903 DOI: 10.1242/dev.112979] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The development of the central nervous system is known to result from two sequential events. First, an inductive event of the mesoderm on the overlying ectoderm that generates a neural plate that, after rolling into a neural tube, acts as the main source of neural progenitors. Second, the axial regionalization of the neural plate that will result in the specification of neurons with different anteroposterior identities. Although this description of the process applies with ease to amphibians and fish, it is more difficult to confirm in amniote embryos. Here, a specialized population of cells emerges at the end of gastrulation that, under the influence of Wnt and FGF signalling, expands and generates the spinal cord and the paraxial mesoderm. This population is known as the long-term neuromesodermal precursor (NMp). Here, we show that controlled increases of Wnt/β-catenin and FGF signalling during adherent culture differentiation of mouse embryonic stem cells (mESCs) generates a population with many of the properties of the NMp. A single-cell analysis of gene expression within this population reveals signatures that are characteristic of stem cell populations. Furthermore, when this activation is triggered in three-dimensional aggregates of mESCs, the population self-organizes macroscopically and undergoes growth and axial elongation that mimics some of the features of the embryonic spinal cord and paraxial mesoderm. We use both adherent and three-dimensional cultures of mESCs to probe the establishment and maintenance of NMps and their differentiation.
Collapse
Affiliation(s)
- David A Turner
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | | | | | - Pau Rué
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | - Rebecca Broome
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | - Fernando Faunes
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | | |
Collapse
|
147
|
Abstract
Retrospective clonal analysis in the mouse has demonstrated that the posterior spinal cord neurectoderm and paraxial mesoderm share a common bipotent progenitor. These neuromesodermal progenitors (NMPs) are the source of new axial structures during embryonic rostrocaudal axis elongation and are marked by the simultaneous co-expression of the transcription factors T(Brachyury) (T(Bra)) and Sox2. NMP-like cells have recently been derived from pluripotent stem cells in vitro following combined stimulation of Wnt and fibroblast growth factor (FGF) signaling. Under these conditions the majority of cultures consist of T(Bra)/Sox2 co-expressing cells after 48-72 hours of differentiation. Although the capacity of these cells to generate posterior neural and paraxial mesoderm derivatives has been demonstrated at the population level, it is unknown whether a single in vitro-derived NMP can give rise to both neural and mesodermal cells. Here we demonstrate that T(Bra) positive cells obtained from mouse epiblast stem cells (EpiSCs) after culture in NMP-inducing conditions can generate both neural and mesodermal clones. This finding suggests that, similar to their embryonic counterparts, in vitro-derived NMPs are truly bipotent and can thus be exploited as a model for studying the molecular basis of developmental cell fate decisions.
Collapse
Affiliation(s)
- Anestis Tsakiridis
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Valerie Wilson
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, EH16 4UU, UK
| |
Collapse
|
148
|
Tsakiridis A, Wilson V. Assessing the bipotency of in vitro-derived neuromesodermal progenitors. F1000Res 2015; 4:100. [PMID: 26401264 PMCID: PMC4566282 DOI: 10.12688/f1000research.6345.1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/15/2015] [Indexed: 11/20/2022] Open
Abstract
Retrospective clonal analysis in the mouse has demonstrated that the posterior spinal cord neurectoderm and paraxial mesoderm share a common bipotent progenitor. These neuromesodermal progenitors (NMPs) are the source of new axial structures during embryonic rostrocaudal axis elongation and are marked by the simultaneous co-expression of the transcription factors T(Brachyury) (T(Bra)) and Sox2. NMP-like cells have recently been derived from pluripotent stem cells in vitro following combined stimulation of Wnt and fibroblast growth factor (FGF) signaling. Under these conditions the majority of cultures consist of T(Bra)/Sox2 co-expressing cells after 48-72 hours of differentiation. Although the capacity of these cells to generate posterior neural and paraxial mesoderm derivatives has been demonstrated at the population level, it is unknown whether a single in vitro-derived NMP can give rise to both neural and mesodermal cells. Here we demonstrate that T(Bra) positive cells obtained from mouse epiblast stem cells (EpiSCs) after culture in NMP-inducing conditions can generate both neural and mesodermal clones. This finding suggests that, similar to their embryonic counterparts, in vitro-derived NMPs are truly bipotent and can thus be exploited as a model for studying the molecular basis of developmental cell fate decisions.
Collapse
Affiliation(s)
- Anestis Tsakiridis
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Valerie Wilson
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, Edinburgh, EH16 4UU, UK
| |
Collapse
|
149
|
Sugiyama M, Saitou T, Kurokawa H, Sakaue-Sawano A, Imamura T, Miyawaki A, Iimura T. Live imaging-based model selection reveals periodic regulation of the stochastic G1/S phase transition in vertebrate axial development. PLoS Comput Biol 2014; 10:e1003957. [PMID: 25474567 PMCID: PMC4256085 DOI: 10.1371/journal.pcbi.1003957] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 09/30/2014] [Indexed: 01/07/2023] Open
Abstract
In multicellular organism development, a stochastic cellular response is observed, even when a population of cells is exposed to the same environmental conditions. Retrieving the spatiotemporal regulatory mode hidden in the heterogeneous cellular behavior is a challenging task. The G1/S transition observed in cell cycle progression is a highly stochastic process. By taking advantage of a fluorescence cell cycle indicator, Fucci technology, we aimed to unveil a hidden regulatory mode of cell cycle progression in developing zebrafish. Fluorescence live imaging of Cecyil, a zebrafish line genetically expressing Fucci, demonstrated that newly formed notochordal cells from the posterior tip of the embryonic mesoderm exhibited the red (G1) fluorescence signal in the developing notochord. Prior to their initial vacuolation, these cells showed a fluorescence color switch from red to green, indicating G1/S transitions. This G1/S transition did not occur in a synchronous manner, but rather exhibited a stochastic process, since a mixed population of red and green cells was always inserted between newly formed red (G1) notochordal cells and vacuolating green cells. We termed this mixed population of notochordal cells, the G1/S transition window. We first performed quantitative analyses of live imaging data and a numerical estimation of the probability of the G1/S transition, which demonstrated the existence of a posteriorly traveling regulatory wave of the G1/S transition window. To obtain a better understanding of this regulatory mode, we constructed a mathematical model and performed a model selection by comparing the results obtained from the models with those from the experimental data. Our analyses demonstrated that the stochastic G1/S transition window in the notochord travels posteriorly in a periodic fashion, with doubled the periodicity of the neighboring paraxial mesoderm segmentation. This approach may have implications for the characterization of the pathophysiological tissue growth mode. Cell cycle progression is considered to involve a cellular time-counting machinery for proper morphogenesis and patterning of tissues. Therefore, it is important to understand the regulatory mode of cell cycle progression during physiological and pathological tissue growth, which will benefit tissue engineering therapy and tumor diagnosis. Since cell cycle progression is a highly variable process, it is a challenging task to retrieve the spatiotemporal regulatory mode hidden in heterogeneous cellular behavior. To overcome this issue, we took advantage of live imaging analyses with a fluorescence cell cycle indicator, Fucci technology, and mathematical modeling of developing zebrafish fish embryo as a model system of growing tissue. Our result demonstrated that the developmental growth of embryonic axis progressed in a rhythmic fashion. The presented analyses will benefit the characterization of the regulatory mode of tissue growth, in both physiological and pathological development, such as that involving tumor formation, thus may contribute to cancer diagnosis.
Collapse
Affiliation(s)
- Mayu Sugiyama
- Laboratory for Cell Function and Dynamics, Advanced Technology Development Group, Brain Science Institute, RIKEN, Wako-city, Saitama, Japan
| | - Takashi Saitou
- Department of Molecular Medicine for Pathogenesis, Graduate School of Medicine, Ehime University, Shitsukawa, Toon-city, Ehime, Japan
| | - Hiroshi Kurokawa
- Laboratory for Cell Function and Dynamics, Advanced Technology Development Group, Brain Science Institute, RIKEN, Wako-city, Saitama, Japan
| | - Asako Sakaue-Sawano
- Laboratory for Cell Function and Dynamics, Advanced Technology Development Group, Brain Science Institute, RIKEN, Wako-city, Saitama, Japan
| | - Takeshi Imamura
- Department of Molecular Medicine for Pathogenesis, Graduate School of Medicine, Ehime University, Shitsukawa, Toon-city, Ehime, Japan
- Division of Bio-Imaging, Proteo-Science Center (PROS), Ehime University, Shitsukawa, Toon-city, Ehime, Japan
- Translational Research Center, Ehime University Hospital, Shitsukawa, Toon-city, Ehime, Japan
- * E-mail: (TIm); (AM); (TIi)
| | - Atsushi Miyawaki
- Laboratory for Cell Function and Dynamics, Advanced Technology Development Group, Brain Science Institute, RIKEN, Wako-city, Saitama, Japan
- * E-mail: (TIm); (AM); (TIi)
| | - Tadahiro Iimura
- Laboratory for Cell Function and Dynamics, Advanced Technology Development Group, Brain Science Institute, RIKEN, Wako-city, Saitama, Japan
- Division of Bio-Imaging, Proteo-Science Center (PROS), Ehime University, Shitsukawa, Toon-city, Ehime, Japan
- Translational Research Center, Ehime University Hospital, Shitsukawa, Toon-city, Ehime, Japan
- * E-mail: (TIm); (AM); (TIi)
| |
Collapse
|
150
|
Taguchi A, Nishinakamura R. Nephron reconstitution from pluripotent stem cells. Kidney Int 2014; 87:894-900. [PMID: 25469851 DOI: 10.1038/ki.2014.358] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 05/21/2014] [Accepted: 05/22/2014] [Indexed: 12/17/2022]
Abstract
It has been a challenge in developmental biology and regenerative medicine to generate nephron progenitors that reconstitute the three-dimensional (3D) nephron structure in vitro. Many studies have tried to induce nephron progenitors from pluripotent stem cells by mimicking the developmental processes in vivo. However, the current developmental model does not precisely address the spatiotemporal origin of nephron progenitors, hampering our understanding of cell fate decisions in the kidney. Here, we present a revised model of early-stage kidney specification, suggesting distinct origins of the two major kidney components: the ureteric bud and metanephric mesenchyme. This model enables the induction of metanephric nephron progenitors from both mouse and human pluripotent stem cells. The induced cells self-organize in the presence of Wnt signaling and reconstitute 3D nephron structures including both nephric tubules with a clear lumina and glomeruli with podocytes. The engrafted kidney tissue develops vascularized glomeruli and nephric tubules, but it does not produce urine, suggesting the requirement for further maturation. Nevertheless, the generation of nephron components from human-induced pluripotent stem cells will be useful for future application in regenerative therapy and modeling of congenital kidney diseases in vitro. This review discusses the possibility of de novo organogenesis of a functional kidney from pluripotent stem cells and the future direction toward clinical applications.
Collapse
Affiliation(s)
- Atsuhiro Taguchi
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| |
Collapse
|