1
|
Toh K, Saunders D, Verd B, Steventon B. Zebrafish neuromesodermal progenitors undergo a critical state transition in vivo. iScience 2022; 25:105216. [PMID: 36274939 PMCID: PMC9579027 DOI: 10.1016/j.isci.2022.105216] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 08/05/2022] [Accepted: 09/22/2022] [Indexed: 11/30/2022] Open
Abstract
The transition state model of cell differentiation proposes that a transient window of gene expression stochasticity precedes entry into a differentiated state. Here, we assess this theoretical model in zebrafish neuromesodermal progenitors (NMps) in vivo during late somitogenesis stages. We observed an increase in gene expression variability at the 24 somite stage (24ss) before their differentiation into spinal cord and paraxial mesoderm. Analysis of a published 18ss scRNA-seq dataset showed that the NMp population is noisier than its derivatives. By building in silico composite gene expression maps from image data, we assigned an 'NM index' to in silico NMps based on the expression of neural and mesodermal markers and demonstrated that cell population heterogeneity peaked at 24ss. Further examination revealed cells with gene expression profiles incongruent with their prospective fate. Taken together, our work supports the transition state model within an endogenous cell fate decision making event.
Collapse
Affiliation(s)
- Kane Toh
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | - Dillan Saunders
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | - Berta Verd
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
- Department of Zoology, University of Oxford, Oxford OX1 3SZ, UK
| | | |
Collapse
|
2
|
Van der Mude A. Structure encoding in DNA. J Theor Biol 2020; 492:110205. [PMID: 32070719 DOI: 10.1016/j.jtbi.2020.110205] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 12/29/2019] [Accepted: 02/14/2020] [Indexed: 12/21/2022]
Abstract
It is proposed that transposons and related long non-coding RNA define the fine structure of body parts. Although morphogens have long been known to direct the formation of many gross structures in early embryonic development, they do not have the necessary precision to define a structure down to the individual cellular level. Using the distinction between procedural and declarative knowledge in information processing as an analogy, it is hypothesized that DNA encodes fine structure in a manner that is different from the genetic code for proteins. The hypothesis states that repeated or near-repeated sequences that are in transposons and non-coding RNA define body part structures. As the cells in a body part go through the epigenetic process of differentiation, the action of methylation serves to inactivate all but the relevant structure definitions and some associated cell type genes. The transposons left active will then physically modify the DNA sequence in the heterochromatin to establish the local context in the three-dimensional body part structure. This brings the encoded definition of the cell type to the histone. The histone code for that cell type starts the regulatory cascade that turns on the genes associated with that particular type of cell, transforming it from a multipotent cell to a fully differentiated cell. This mechanism creates structures in the musculoskeletal system, the organs of the body, the major parts of the brain, and other systems.
Collapse
|
3
|
Gallivan CP, Ren H, Read EL. Analysis of Single-Cell Gene Pair Coexpression Landscapes by Stochastic Kinetic Modeling Reveals Gene-Pair Interactions in Development. Front Genet 2020; 10:1387. [PMID: 32082359 PMCID: PMC7005996 DOI: 10.3389/fgene.2019.01387] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/18/2019] [Indexed: 12/04/2022] Open
Abstract
Single-cell transcriptomics is advancing discovery of the molecular determinants of cell identity, while spurring development of novel data analysis methods. Stochastic mathematical models of gene regulatory networks help unravel the dynamic, molecular mechanisms underlying cell-to-cell heterogeneity, and can thus aid interpretation of heterogeneous cell-states revealed by single-cell measurements. However, integrating stochastic gene network models with single cell data is challenging. Here, we present a method for analyzing single-cell gene-pair coexpression patterns, based on biophysical models of stochastic gene expression and interaction dynamics. We first developed a high-computational-throughput approach to stochastic modeling of gene-pair coexpression landscapes, based on numerical solution of gene network Master Equations. We then comprehensively catalogued coexpression patterns arising from tens of thousands of gene-gene interaction models with different biochemical kinetic parameters and regulatory interactions. From the computed landscapes, we obtain a low-dimensional "shape-space" describing distinct types of coexpression patterns. We applied the theoretical results to analysis of published single cell RNA sequencing data and uncovered complex dynamics of coexpression among gene pairs during embryonic development. Our approach provides a generalizable framework for inferring evolution of gene-gene interactions during critical cell-state transitions.
Collapse
Affiliation(s)
- Cameron P. Gallivan
- Department of Chemical & Biomolecular Engineering, University of California, Irvine, CA, United States
| | - Honglei Ren
- NSF-Simons Center for Multiscale Cell Fate, University of California, Irvine, CA, United States
- Mathematical and Computational Systems Biology Graduate Program, University of California, Irvine, CA, United States
| | - Elizabeth L. Read
- Department of Chemical & Biomolecular Engineering, University of California, Irvine, CA, United States
- NSF-Simons Center for Multiscale Cell Fate, University of California, Irvine, CA, United States
| |
Collapse
|
4
|
Favarolo MB, López SL. Notch signaling in the division of germ layers in bilaterian embryos. Mech Dev 2018; 154:122-144. [PMID: 29940277 DOI: 10.1016/j.mod.2018.06.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 06/08/2018] [Accepted: 06/18/2018] [Indexed: 01/09/2023]
Abstract
Bilaterian embryos are triploblastic organisms which develop three complete germ layers (ectoderm, mesoderm, and endoderm). While the ectoderm develops mainly from the animal hemisphere, there is diversity in the location from where the endoderm and the mesoderm arise in relation to the animal-vegetal axis, ranging from endoderm being specified between the ectoderm and mesoderm in echinoderms, and the mesoderm being specified between the ectoderm and the endoderm in vertebrates. A common feature is that part of the mesoderm segregates from an ancient bipotential endomesodermal domain. The process of segregation is noisy during the initial steps but it is gradually refined. In this review, we discuss the role of the Notch pathway in the establishment and refinement of boundaries between germ layers in bilaterians, with special focus on its interaction with the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- María Belén Favarolo
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Biología Celular y Neurociencias "Prof. E. De Robertis" (IBCN), Facultad de Medicina, Laboratorio de Embriología Molecular "Prof. Dr. Andrés E. Carrasco", Buenos Aires, Argentina
| | - Silvia L López
- Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Biología Celular y Neurociencias "Prof. E. De Robertis" (IBCN), Facultad de Medicina, Laboratorio de Embriología Molecular "Prof. Dr. Andrés E. Carrasco", Buenos Aires, Argentina.
| |
Collapse
|
5
|
Briggs JA, Weinreb C, Wagner DE, Megason S, Peshkin L, Kirschner MW, Klein AM. The dynamics of gene expression in vertebrate embryogenesis at single-cell resolution. Science 2018; 360:science.aar5780. [PMID: 29700227 DOI: 10.1126/science.aar5780] [Citation(s) in RCA: 374] [Impact Index Per Article: 53.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 04/16/2018] [Indexed: 12/11/2022]
Abstract
Time series of single-cell transcriptome measurements can reveal dynamic features of cell differentiation pathways. From measurements of whole frog embryos spanning zygotic genome activation through early organogenesis, we derived a detailed catalog of cell states in vertebrate development and a map of differentiation across all lineages over time. The inferred map recapitulates most if not all developmental relationships and associates new regulators and marker genes with each cell state. We find that many embryonic cell states appear earlier than previously appreciated. We also assess conflicting models of neural crest development. Incorporating a matched time series of zebrafish development from a companion paper, we reveal conserved and divergent features of vertebrate early developmental gene expression programs.
Collapse
Affiliation(s)
- James A Briggs
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Caleb Weinreb
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Daniel E Wagner
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Sean Megason
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Leonid Peshkin
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Marc W Kirschner
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA.
| | - Allon M Klein
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
6
|
Abstract
DNA methylation is a major epigenetic modification of vertebrate genomes that is mostly associated with transcriptional repression. During embryogenesis, DNA methylation together with other epigenetic factors plays an essential role in selecting and maintaining cell identity. Recent technological advances are now allowing for the exploration of this mark at unprecedented resolution. This has resulted in a wealth of studies describing the developmental roles of DNA methylation in various vertebrate model systems. It is now evident that in certain contexts DNA methylation can act as a key regulator of cell identity establishment, whereas in many other cases the quantity of DNA methylation will merely reflect other upstream regulatory changes. For example, a number of studies have indicated that DNA methylation might be dispensable for pluripotency stages of embryonic development. Nevertheless, targeted deposition and removal of DNA methylation by DNMTs and TET proteins, respectively, appears to be required for vertebrate gastrulation. Here we review the roles of DNA methylation in the establishment and maintenance of cell identity during development, with a special emphasis on insights obtained from in vivo studies.
Collapse
|
7
|
Brg1 chromatin remodeling ATPase balances germ layer patterning by amplifying the transcriptional burst at midblastula transition. PLoS Genet 2017; 13:e1006757. [PMID: 28498870 PMCID: PMC5428918 DOI: 10.1371/journal.pgen.1006757] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 04/11/2017] [Indexed: 12/21/2022] Open
Abstract
Zygotic gene expression programs control cell differentiation in vertebrate development. In Xenopus, these programs are initiated by local induction of regulatory genes through maternal signaling activities in the wake of zygotic genome activation (ZGA) at the midblastula transition (MBT). These programs lay down the vertebrate body plan through gastrulation and neurulation, and are accompanied by massive changes in chromatin structure, which increasingly constrain cellular plasticity. Here we report on developmental functions for Brahma related gene 1 (Brg1), a key component of embyronic SWI/SNF chromatin remodeling complexes. Carefully controlled, global Brg1 protein depletion in X. tropicalis and X. laevis causes embryonic lethality or developmental arrest from gastrulation on. Transcriptome analysis at late blastula, before development becomes arrested, indicates predominantly a role for Brg1 in transcriptional activation of a limited set of genes involved in pattern specification processes and nervous system development. Mosaic analysis by targeted microinjection defines Brg1 as an essential amplifier of gene expression in dorsal (BCNE/Nieuwkoop Center) and ventral (BMP/Vent) signaling centers. Moreover, Brg1 is required and sufficient for initiating axial patterning in cooperation with maternal Wnt signaling. In search for a common denominator of Brg1 impact on development, we have quantitatively filtered global mRNA fluctuations at MBT. The results indicate that Brg1 is predominantly required for genes with the highest burst of transcriptional activity. Since this group contains many key developmental regulators, we propose Brg1 to be responsible for raising their expression above threshold levels in preparation for embryonic patterning. Brahma-related-gene-1 (Brg1) is a catalytic subunit of mammalian SWI/SNF chromatin remodeling complexes. Loss of maternal Brg1 protein arrests development in mice at the 2-cell stage, while null homozygotes die at the blastocyst stage. These early requirements have precluded any analysis of Brg1’s embryonic functions. Here we present data from X. laevis and X. tropicalis, which for the first time describe a role for Brg1 during germ layer patterning and axis formation. Brg1-depleted embryos fail to develop past gastrulation. Genome-wide transcriptome analysis at late blastula stage, before the developmental arrest, shows that Brg1 is required predominantly for transcriptional activation of a limited set of genes involved in pattern specification processes and nervous system development shortly after midblastula transition. Mosaic analysis by targeted microinjection defines Brg1 as an essential amplifier of gene expression in dorsal (BCNE and Nieuwkoop center) and ventral (BMP/Vent) signaling centers, being required and sufficient to initiate axial patterning by cooperating with canonical Wnt signaling. Since Brg1-dependent genes share a high burst of transcriptional activation before gastrulation, we propose a systemic role for Brg1 as transcriptional amplifier, which balances the embryonic patterning process.
Collapse
|
8
|
Flickinger R. AT-rich repetitive DNA sequences, transcription frequency and germ layer determination. Mech Dev 2015; 138 Pt 3:227-32. [PMID: 26506258 DOI: 10.1016/j.mod.2015.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 10/19/2015] [Accepted: 10/21/2015] [Indexed: 01/30/2023]
Abstract
Non-coding sequences of frog embryo endoderm poly (A+) nuclear RNA are AU-enriched, as compared to those of ectoderm and mesoderm. Endoderm blastomeres contain much less H1 histone than is present in ectoderm and mesoderm. H1 histone preferentially binds AT-rich DNA sequences to repress their transcription. The AT-enrichment of non-coding DNA sequences transcribed into poly (A+) nuclear RNA, as well as the low amount of H1 histone, may contribute to the higher transcription frequency of mRNA of endoderm, as compared to that of ectoderm and mesoderm. A greater accumulation of H1 histone in presumptive mesoderm and ectoderm may prevent transcription of endoderm specifying genes in mesoderm and ectoderm. Experimental upregulation of various transcription factors (TFs) can redirect germ layer fate. Most of these TFs bind AT-rich consensus sequences in DNA, suggesting that H1 histone and TFs active during germ layer determination are binding similar sequences.
Collapse
Affiliation(s)
- Reed Flickinger
- Emeritus Department, Biological Sciences State University of New York at Buffalo, Buffalo, N.Y. 14260, USA.
| |
Collapse
|
9
|
Klein AM, Mazutis L, Akartuna I, Tallapragada N, Veres A, Li V, Peshkin L, Weitz DA, Kirschner MW. Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells. Cell 2015; 161:1187-1201. [PMID: 26000487 PMCID: PMC4441768 DOI: 10.1016/j.cell.2015.04.044] [Citation(s) in RCA: 2281] [Impact Index Per Article: 228.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Revised: 02/23/2015] [Accepted: 04/20/2015] [Indexed: 12/11/2022]
Abstract
It has long been the dream of biologists to map gene expression at the single-cell level. With such data one might track heterogeneous cell sub-populations, and infer regulatory relationships between genes and pathways. Recently, RNA sequencing has achieved single-cell resolution. What is limiting is an effective way to routinely isolate and process large numbers of individual cells for quantitative in-depth sequencing. We have developed a high-throughput droplet-microfluidic approach for barcoding the RNA from thousands of individual cells for subsequent analysis by next-generation sequencing. The method shows a surprisingly low noise profile and is readily adaptable to other sequencing-based assays. We analyzed mouse embryonic stem cells, revealing in detail the population structure and the heterogeneous onset of differentiation after leukemia inhibitory factor (LIF) withdrawal. The reproducibility of these high-throughput single-cell data allowed us to deconstruct cell populations and infer gene expression relationships. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Allon M Klein
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Linas Mazutis
- School of Engineering and Applied Sciences (SEAS), Harvard University, Cambridge, MA 02138, USA; Vilnius University Institute of Biotechnology, Vilnius LT-02241, Lithuania
| | - Ilke Akartuna
- School of Engineering and Applied Sciences (SEAS), Harvard University, Cambridge, MA 02138, USA
| | - Naren Tallapragada
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Adrian Veres
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Victor Li
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Leonid Peshkin
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - David A Weitz
- School of Engineering and Applied Sciences (SEAS), Harvard University, Cambridge, MA 02138, USA.
| | - Marc W Kirschner
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
10
|
Dhorne-Pollet S, Thélie A, Pollet N. Validation of novel reference genes for RT-qPCR studies of gene expression in Xenopus tropicalis during embryonic and post-embryonic development. Dev Dyn 2013; 242:709-17. [PMID: 23559567 DOI: 10.1002/dvdy.23972] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Revised: 02/04/2013] [Accepted: 03/26/2013] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Accurate interpretation of transcriptome profiling by quantitative PCR requires the establishment of species-specific standards. However, the selection of reference genes for assessing RNA expression profiles in Xenopus laevis and Xenopus tropicalis was mostly based on historical reasons and they often only reflect the traditions of a laboratory. RESULTS We investigated the expression stability of 10 genes (dicer1, drosha, eef1a1, elavl3, gsc, h4, odc1, rpl8, smn2, tbp), 8 of which are commonly used as internal controls in published RT-qPCR experiments. We defined specific primer pairs and evaluated their suitability as reference genes by performing RT-qPCR expression profiling in Xenopus tropicalis. Gene expression stability was assayed in a set of 15 developmental stages from the egg to the froglet, and in dissected embryos. CONCLUSIONS Overall, we determined a set of qualified reference genes for distinct developmental periods. We recommend the use of dicer1, drosha, eef1a1, and smn2 from early embryonic development up to the end of metamorphosis. During early embryogenesis drosha, eef1a1, smn2 are suitable. For the whole post-embryonic development and for metamorphic stages including pro-metamorphosis and metamorphic climax, we recommend the use of drosha and smn2. These reference genes should prove their usefulness for data comparison across studies.
Collapse
|
11
|
Zhao J, Lambert G, Meijer AH, Rosa FM. The transcription factor Vox represses endoderm development by interacting with Casanova and Pou2. Development 2013; 140:1090-9. [DOI: 10.1242/dev.082008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Endoderm and mesoderm are both formed upon activation of Nodal signaling but how endoderm differentiates from mesoderm is still poorly explored. The sox-related gene casanova (sox32) acts downstream of the Nodal signal, is essential for endoderm development and requires the co-factor Pou2 (Pou5f1, Oct3, Oct4) in this process. Conversely, BMP signals have been shown to inhibit endoderm development by an as yet unexplained mechanism. In a search for Casanova regulators in zebrafish, we identified two of its binding partners as the transcription factors Pou2 and Vox, a member of the Vent group of proteins also involved in the patterning of the gastrula. In overexpression studies we show that vox and/or Vent group genes inhibit the capacity of Casanova to induce endoderm, even in the presence of its co-factor Pou2, and that Vox acts as a repressor in this process. We further show that vox, but not other members of the Vent group, is essential for defining the proper endodermal domain size at gastrulation. In this process, vox acts downstream of BMPs. Cell fate analysis further shows that Vox plays a key role downstream of BMP signals in regulating the capacity of Nodal to induce endoderm versus mesoderm by modulating the activity of the Casanova/Pou2 regulatory system.
Collapse
Affiliation(s)
- Jue Zhao
- INSERM U1024, F-75005 Paris, France
- CNRS UMR 8197, F-75005 Paris, France
- IBENS, Institut de Biologie de l’Ecole Normale Supérieure, F-75230 Paris, France
- College of Life Sciences, Peking University, Beijing 100871, P. R. China
| | - Guillaume Lambert
- INSERM U1024, F-75005 Paris, France
- CNRS UMR 8197, F-75005 Paris, France
- IBENS, Institut de Biologie de l’Ecole Normale Supérieure, F-75230 Paris, France
| | | | - Frederic M. Rosa
- INSERM U1024, F-75005 Paris, France
- CNRS UMR 8197, F-75005 Paris, France
- IBENS, Institut de Biologie de l’Ecole Normale Supérieure, F-75230 Paris, France
| |
Collapse
|
12
|
Cerdan C, McIntyre BAS, Mechael R, Levadoux-Martin M, Yang J, Lee JB, Bhatia M. Activin A promotes hematopoietic fated mesoderm development through upregulation of brachyury in human embryonic stem cells. Stem Cells Dev 2012; 21:2866-77. [PMID: 22548442 DOI: 10.1089/scd.2012.0053] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The development of the hematopoietic system involves multiple cellular steps beginning with the formation of the mesoderm from the primitive streak, followed by emergence of precursor populations that become committed to either the endothelial or hematopoietic lineages. A number of growth factors such as activins and fibroblast growth factors (FGFs) are known to regulate the early specification of hematopoietic fated mesoderm, notably in amphibians. However, the potential roles of these factors in the development of mesoderm and subsequent hematopoiesis in the human have yet to be delineated. Defining the cellular and molecular mechanisms by which combinations of mesoderm-inducing factors regulate this stepwise process in human cells in vitro is central to effectively directing human embryonic stem cell (hESC) hematopoietic differentiation. Herein, using hESC-derived embryoid bodies (EBs), we show that Activin A, but not basic FGF/FGF2 (bFGF), promotes hematopoietic fated mesodermal specification from pluripotent human cells. The effect of Activin A treatment relies on the presence of bone morphogenetic protein 4 (BMP4) and both of the hematopoietic cytokines stem cell factor and fms-like tyrosine kinase receptor-3 ligand, and is the consequence of 2 separate mechanisms occurring at 2 different stages of human EB development from mesoderm to blood. While Activin A promotes the induction of mesoderm, as indicated by the upregulation of Brachyury expression, which represents the mesodermal precursor required for hematopoietic development, it also contributes to the expansion of cells already committed to a hematopoietic fate. As hematopoietic development requires the transition through a Brachyury+ intermediate, we demonstrate that hematopoiesis in hESCs is impaired by the downregulation of Brachyury, but is unaffected by its overexpression. These results demonstrate, for the first time, the functional significance of Brachyury in the developmental program of hematopoietic differentiation from hESCs and provide an in-depth understanding of the molecular cues that orchestrate stepwise development of hematopoiesis in a human system.
Collapse
Affiliation(s)
- Chantal Cerdan
- McMaster Stem Cell and Cancer Research Institute, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
13
|
Bogdanović O, van Heeringen SJ, Veenstra GJC. The epigenome in early vertebrate development. Genesis 2012; 50:192-206. [PMID: 22139962 PMCID: PMC3294079 DOI: 10.1002/dvg.20831] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Revised: 11/22/2011] [Accepted: 11/23/2011] [Indexed: 01/04/2023]
Abstract
Epigenetic regulation defines the commitment and potential of cells, including the limitations in their competence to respond to inducing signals. This review discusses the developmental origins of chromatin state in Xenopus and other vertebrate species and provides an overview of its use in genome annotation. In most metazoans the embryonic genome is transcriptionally quiescent after fertilization. This involves nucleosome-dense chromatin, repressors and a temporal deficiency in the transcription machinery. Active histone modifications such as H3K4me3 appear in pluripotent blastula embryos, whereas repressive marks such as H3K27me3 show a major increase in enrichment during late blastula and gastrula stages. The H3K27me3 modification set by Polycomb restricts ectopic lineage-specific gene expression. Pluripotent chromatin in Xenopus embryos is relatively unconstrained, whereas the pluripotent cell lineage in mammalian embryos harbors a more enforced type of pluripotent chromatin.
Collapse
Affiliation(s)
- Ozren Bogdanović
- Radboud University Nijmegen, Dept. Molecular Biology, Faculty of Science, Nijmegen Centre of Molecular Life Sciences, Nijmegen, The Netherlands
| | - Simon J. van Heeringen
- Radboud University Nijmegen, Dept. Molecular Biology, Faculty of Science, Nijmegen Centre of Molecular Life Sciences, Nijmegen, The Netherlands
| | - Gert Jan C. Veenstra
- Radboud University Nijmegen, Dept. Molecular Biology, Faculty of Science, Nijmegen Centre of Molecular Life Sciences, Nijmegen, The Netherlands
| |
Collapse
|
14
|
Schneider TD, Arteaga-Salas JM, Mentele E, David R, Nicetto D, Imhof A, Rupp RAW. Stage-specific histone modification profiles reveal global transitions in the Xenopus embryonic epigenome. PLoS One 2011; 6:e22548. [PMID: 21814581 PMCID: PMC3142184 DOI: 10.1371/journal.pone.0022548] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Accepted: 06/29/2011] [Indexed: 02/03/2023] Open
Abstract
Vertebrate embryos are derived from a transitory pool of pluripotent cells. By the process of embryonic induction, these precursor cells are assigned to specific fates and differentiation programs. Histone post-translational modifications are thought to play a key role in the establishment and maintenance of stable gene expression patterns underlying these processes. While on gene level histone modifications are known to change during differentiation, very little is known about the quantitative fluctuations in bulk histone modifications during development. To investigate this issue we analysed histones isolated from four different developmental stages of Xenopus laevis by mass spectrometry. In toto, we quantified 59 modification states on core histones H3 and H4 from blastula to tadpole stages. During this developmental period, we observed in general an increase in the unmodified states, and a shift from histone modifications associated with transcriptional activity to transcriptionally repressive histone marks. We also compared these naturally occurring patterns with the histone modifications of murine ES cells, detecting large differences in the methylation patterns of histone H3 lysines 27 and 36 between pluripotent ES cells and pluripotent cells from Xenopus blastulae. By combining all detected modification transitions we could cluster their patterns according to their embryonic origin, defining specific histone modification profiles (HMPs) for each developmental stage. To our knowledge, this data set represents the first compendium of covalent histone modifications and their quantitative flux during normogenesis in a vertebrate model organism. The HMPs indicate a stepwise maturation of the embryonic epigenome, which may be causal to the progressing restriction of cellular potency during development.
Collapse
Affiliation(s)
- Tobias D Schneider
- Department of Molecular Biology, Adolf-Butenandt Institut, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | | | | | | | | | | |
Collapse
|
15
|
Klymkowsky MW, Rossi CC, Artinger KB. Mechanisms driving neural crest induction and migration in the zebrafish and Xenopus laevis. Cell Adh Migr 2010; 4:595-608. [PMID: 20962584 PMCID: PMC3011258 DOI: 10.4161/cam.4.4.12962] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Accepted: 07/09/2010] [Indexed: 01/09/2023] Open
Abstract
The neural crest is an evolutionary adaptation, with roots in the formation of mesoderm. Modification of neural crest behavior has been is critical for the evolutionary diversification of the vertebrates and defects in neural crest underlie a range of human birth defects. There has been a tremendous increase in our knowledge of the molecular, cellular, and inductive interactions that converge on defining the neural crest and determining its behavior. While there is a temptation to look for simple models to explain neural crest behavior, the reality is that the system is complex in its circuitry. In this review, our goal is to identify the broad features of neural crest origins (developmentally) and migration (cellularly) using data from the zebrafish (teleost) and Xenopus laevis (tetrapod amphibian) in order to illuminate where general mechanisms appear to be in play, and equally importantly, where disparities in experimental results suggest areas of profitable study.
Collapse
Affiliation(s)
- Michael W Klymkowsky
- Department of Molecular, Cellular and Developmental Biology; University of Colorado Boulder; Boulder, CO USA
| | - Christy Cortez Rossi
- Department of Craniofacial Biology; University of Colorado Denver; School of Dental Medicine; Aurora, CO USA
| | - Kristin Bruk Artinger
- Department of Craniofacial Biology; University of Colorado Denver; School of Dental Medicine; Aurora, CO USA
| |
Collapse
|
16
|
Delta-Notch signaling is involved in the segregation of the three germ layers in Xenopus laevis. Dev Biol 2010; 339:477-92. [PMID: 20079726 DOI: 10.1016/j.ydbio.2010.01.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2009] [Revised: 01/06/2010] [Accepted: 01/07/2010] [Indexed: 11/20/2022]
Abstract
In vertebrates, the induction of the three germ layers (ectoderm, mesoderm and endoderm) has been extensively studied, but less is known about how they segregate. Here, we investigated whether Delta-Notch signaling is involved in this process. Activating the pathway in the marginal zone with Notch(ICD) resulted in an expansion of endodermal and neural ectoderm precursors, leaving a thinner mesodermal ring around the blastopore at gastrula stage, when germ layers are segregated. On the other hand, when the pathway was blocked with Delta-1(STU) or with an antisense morpholino oligonucleotide against Notch, the pan-mesodermal brachyury (bra) domain was expanded and the neural border was moved animalwards. Strikingly, the suprablastoporal endoderm was either expanded when Delta-1 signaling was blocked, or reduced after the general knock-down of Notch. In addition, either activating or blocking the pathway delays the blastopore closure. We conclude that the process of delimiting the three germ layers requires Notch signaling, which may be finely regulated by ligands and/or involve non-canonical components of the pathway. Moreover, Notch activity must be modulated at appropriate levels during this process in order to keep normal morphogenetic movements during gastrulation.
Collapse
|
17
|
Zhang Q, Zheng K, Ma S, Tong Y, Luo C. Goldfish β-catenin cell-autonomously inhibits the expression of early neural development regulating gene vsx1. CHINESE SCIENCE BULLETIN-CHINESE 2010. [DOI: 10.1007/s11434-009-0714-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
18
|
Kwek J, De Iongh R, Nicholas K, Familari M. Molecular insights into evolution of the vertebrate gut: focus on stomach and parietal cells in the marsupial,Macropus eugenii. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2009; 312:613-24. [DOI: 10.1002/jez.b.21227] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
19
|
Middleton AM, King JR, Loose M. Bistability in a model of mesoderm and anterior mesendoderm specification in Xenopus laevis. J Theor Biol 2009; 260:41-55. [PMID: 19490918 DOI: 10.1016/j.jtbi.2009.05.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2008] [Revised: 04/10/2009] [Accepted: 05/16/2009] [Indexed: 11/20/2022]
Abstract
In this paper we develop a model of mesendoderm specification in Xenopus laevis based on an existing gene regulation network. The mesendoderm is a population of cells that may contribute to either the mesoderm or endoderm. The model that we develop encompasses the time evolution of transcription factor concentrations in a single cell and is shown to have stable steady states that correspond to mesoderm and anterior mesendodermal cell types, but not endoderm (except in cells where Goosecoid expression is inhibited). Both in vitro and in vivo versions of the model are developed and analysed, the former indicating how cell fate is determined in large part by the concentration of Activin administered to a cell, with the model results comparing favourably with current quantitative experimental data. A numerical investigation of the in vivo model suggests that cell fate is determined largely by a VegT and beta-Catenin pre-pattern, subsequently being reinforced by Nodal. We argue that this sensitivity of the model to a VegT and beta-Catenin pre-pattern indicates that a key VegT self-limiting mechanism (for which there is experimental evidence) is absent from the model. Furthermore, we find that the lack of a steady state corresponding to endoderm is entirely consistent with current in vivo data, and that the in vivo model corresponds to mesendoderm specification on the dorsal, but not the ventral, side of the embryo.
Collapse
|
20
|
Halley JD, Burden FR, Winkler DA. Stem cell decision making and critical-like exploratory networks. Stem Cell Res 2009; 2:165-77. [DOI: 10.1016/j.scr.2009.03.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2009] [Revised: 02/24/2009] [Accepted: 03/06/2009] [Indexed: 10/21/2022] Open
|
21
|
|
22
|
Qu XB, Pan J, Zhang C, Huang SY. Sox17 facilitates the differentiation of mouse embryonic stem cells into primitive and definitive endoderm in vitro. Dev Growth Differ 2009; 50:585-93. [PMID: 19238729 DOI: 10.1111/j.1440-169x.2008.01056.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The Sox family of HMG (high mobility group)-box transcription factors are highly conserved in vertebrates. Sox members are involved in various developmental processes. Among them Sox17 has been demonstrated to function as an endoderm determinant in zebrafish and Xenopus, respectively. However, little is known about the role of Sox17 in mouse embryonic stem cell (ESC) differentiation. In our research, we investigated the effect of Sox17 on mouse ESC and embryoid body (EB) differentiation. The results demonstrated that Sox17 overexpression upregulated a set of endoderm-specific gene markers, suggesting that Sox17 overexpression induced an ESC differentiation program towards both primitive and definitive endoderm. We believe this finding brings new insights into the understanding of ESC differentiation and the organogenesis of endodermal derivatives.
Collapse
Affiliation(s)
- Xue-Bin Qu
- Key Laboratory of Animal Resistance Research, College of Life Science, Shandong Normal University, 88 East Wenhua Road, Jinan City, Shandong Province, China
| | | | | | | |
Collapse
|
23
|
Abstract
The endoderm germ layer contributes to the respiratory and gastrointestinal tracts and to all of their associated organs. Over the past decade, studies in vertebrate model organisms, including frog, fish, chick, and mouse, have greatly enhanced our understanding of the molecular basis of endoderm organ development. We review this progress with a focus on early stages of endoderm organogenesis including endoderm formation, gut tube morphogenesis and patterning, and organ specification. Lastly, we discuss how developmental mechanisms that regulate endoderm organogenesis are used to direct differentiation of embryonic stem cells into specific adult cell types, which function to alleviate disease symptoms in animal models.
Collapse
Affiliation(s)
- Aaron M. Zorn
- Division of Developmental Biology, Cincinnati Children’s Research Foundation and Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio 45229
| | - James M. Wells
- Division of Developmental Biology, Cincinnati Children’s Research Foundation and Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio 45229
| |
Collapse
|
24
|
Koop D, Holland LZ. The basal chordate amphioxus as a simple model for elucidating developmental mechanisms in vertebrates. ACTA ACUST UNITED AC 2008; 84:175-87. [DOI: 10.1002/bdrc.20128] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
25
|
Woodland HR, Zorn AM. The core endodermal gene network of vertebrates: combining developmental precision with evolutionary flexibility. Bioessays 2008; 30:757-65. [PMID: 18623060 DOI: 10.1002/bies.20785] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Embryonic development combines paradoxical properties: it has great precision, it is usually conducted at breakneck speed and it is flexible on relatively short evolutionary time scales, particularly at early stages. While these features appear mutually exclusive, we consider how they may be reconciled by the properties of key early regulatory networks. We illustrate these ideas with the network that controls development of endoderm progenitors. We argue that this network enables precision because of its intrinsic stability, self propagation and dependence on signalling. The network enables high developmental speed because it is rapidly established by maternal inputs at multiple points. In turn these properties confer flexibility on an evolutionary time scale because they can be initiated in many ways, while buffering essential progenitor cell populations against changes in their embryonic environment on both evolutionary and developmental time scales. Although stable, these networks must be capable of rapid dissolution as cell differentiation progresses. While we focus on the core early endodermal network of vertebrates, we argue that these properties are likely to be general in early embryonic stem cell populations, such as mammalian ES cells.
Collapse
Affiliation(s)
- Hugh R Woodland
- Department of Biological Sciences, University of Warwick, Coventry, UK.
| | | |
Collapse
|
26
|
Upstream stimulatory factors, USF1 and USF2 are differentially expressed during Xenopus embryonic development. Gene Expr Patterns 2008; 8:376-381. [PMID: 18585979 DOI: 10.1016/j.gep.2008.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2007] [Revised: 05/16/2008] [Accepted: 05/20/2008] [Indexed: 01/22/2023]
Abstract
Upstream stimulatory factors (USF) 1 and 2 are members of the basic helix-loop-helix leucine zipper transcription factor family. They are considered to play critical roles in cell-cycle regulation and chromatin remodeling. Their gene expression patterns are considered ubiquitous but have not been fully investigated in terms of embryogenesis. We examined the expression of the genes encoding USF1 and USF2 in Xenopus laevis during embryonic development. Expression of both genes was first detected as maternal transcripts and was observed continuously throughout development. However, in situ hybridization analysis revealed that the two genes were expressed differentially. In the late blastula, both genes were expressed in the blastocoel roof and marginal zone. At the gastrula stage, USF2 was strongly expressed in the sensorial layer of the ectoderm and in the mesoderm, whereas USF1 expression was hardly detectable. From the neurula stage onward, expression of both genes was markedly enhanced in the neural tissues, neural crest, eye and otic vesicle. However, spatial expression of the genes within the neural tube differed in that the strongest USF1 signals were observed in the lateral region of the basal plate and the strongest USF2 ones in the dorsal region of the neural tube. Expression of the two genes occurred in different mesoderm derivatives at the tailbud stage (USF1, somite; USF2, pronephros and lateral plate mesoderm of the tail region). USF1 was expressed in the notochord of the early neurula, but was lost at the stage.
Collapse
|
27
|
Robertson SH, Smith CK, Langhans AL, McLinden SE, Oberhardt MA, Jakab KR, Dzamba B, DeSimone DW, Papin JA, Peirce SM. Multiscale computational analysis of Xenopus laevis morphogenesis reveals key insights of systems-level behavior. BMC SYSTEMS BIOLOGY 2007; 1:46. [PMID: 17953751 PMCID: PMC2190763 DOI: 10.1186/1752-0509-1-46] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2007] [Accepted: 10/22/2007] [Indexed: 11/25/2022]
Abstract
Background Tissue morphogenesis is a complex process whereby tissue structures self-assemble by the aggregate behaviors of independently acting cells responding to both intracellular and extracellular cues in their environment. During embryonic development, morphogenesis is particularly important for organizing cells into tissues, and although key regulatory events of this process are well studied in isolation, a number of important systems-level questions remain unanswered. This is due, in part, to a lack of integrative tools that enable the coupling of biological phenomena across spatial and temporal scales. Here, we present a new computational framework that integrates intracellular signaling information with multi-cell behaviors in the context of a spatially heterogeneous tissue environment. Results We have developed a computational simulation of mesendoderm migration in the Xenopus laevis explant model, which is a well studied biological model of tissue morphogenesis that recapitulates many features of this process during development in humans. The simulation couples, via a JAVA interface, an ordinary differential equation-based mass action kinetics model to compute intracellular Wnt/β-catenin signaling with an agent-based model of mesendoderm migration across a fibronectin extracellular matrix substrate. The emergent cell behaviors in the simulation suggest the following properties of the system: maintaining the integrity of cell-to-cell contact signals is necessary for preventing fractionation of cells as they move, contact with the Fn substrate and the existence of a Fn gradient provides an extracellular feedback loop that governs migration speed, the incorporation of polarity signals is required for cells to migrate in the same direction, and a delicate balance of integrin and cadherin interactions is needed to reproduce experimentally observed migratory behaviors. Conclusion Our computational framework couples two different spatial scales in biology: intracellular with multicellular. In our simulation, events at one scale have quantitative and dynamic impact on events at the other scale. This integration enables the testing and identification of key systems-level hypotheses regarding how signaling proteins affect overall tissue-level behavior during morphogenesis in an experimentally verifiable system. Applications of this approach extend to the study of tissue patterning processes that occur during adulthood and disease, such as tumorgenesis and atherogenesis.
Collapse
Affiliation(s)
- Scott H Robertson
- Department of Biomedical Engineering, University of Virginia, Box 800759, Charlottesville, VA 22908, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Concentration gradients of small diffusible molecules called morphogens are key regulators of development, specifying position during pattern formation in the embryo. It is now becoming clear that additional or alternative mechanisms involving interactions among cells are also crucial for positional specification.
Collapse
Affiliation(s)
- Michel Kerszberg
- Université Pierre et Marie Curie-Paris 6, UMR7138 CNRS, 75005 Paris, France.
| | | |
Collapse
|
29
|
Howard L, Rex M, Clements D, Woodland HR. Regulation of the Xenopus Xsox17alpha(1) promoter by co-operating VegT and Sox17 sites. Dev Biol 2007; 310:402-15. [PMID: 17719026 PMCID: PMC2098691 DOI: 10.1016/j.ydbio.2007.07.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2007] [Revised: 07/03/2007] [Accepted: 07/23/2007] [Indexed: 11/23/2022]
Abstract
The gene encoding the Sox F-group transcription factor Xsox17α1 is specifically expressed throughout the entire region of the Xenopus blastula fated to become endoderm, and is important in controlling endodermal development. Xsox17α1 is a direct target of the maternal endodermal determinant VegT and of Sox17 itself. We have analysed the promoter of the Xenopus laevis Xsox17α1 gene by transgenesis, and have identified two important control elements which reside about 9 kb upstream at the start of transcription. These elements individually drive transgenic endodermal expression in the blastula and gastrula. One contains functional, cooperating VegT and Sox-binding consensus sites. The Sox sites in this region are occupied in vivo. The other responds to TGF-β signals like Activin or Nodals that act through Smad2/3. We propose that these two regions co-operate in regulating the early endodermal expression of the Xsox17α1 gene.
Collapse
|
30
|
Abstract
Asymmetries in the egg, established during oogenesis, set the stage for a cascade of intercellular signaling events leading to differential gene expression and subsequent tissue and organ formation. Maternally supplied Sox-type transcription factors have recently emerged as key components in the patterning of the early embryo and the regulation of embryonic stem cell differentiation. In deuterostomes, B1-type Soxs are asymmetrically localized to the future animal/ectodermal region where they act to suppress mesendodermal, and favor neuroectodermal differentiation, while vegetally localized F-type Soxs are involved in mesendodermal differentiation. Here, we review past observations and present new data from studies on the clawed frog Xenopus laevis. Animally localized Sox3 acts to inhibit Nodal (Xnr5 and Xnr6) expression, and induces the expression of genes (Ectodermin, Xema, and Coco) whose products repress Nodal signaling. Vegetally localized Sox7 positively regulates Nodal (Xnr4, Xnr5, and Xnr6) expression, as well as the expression of genes involved in mesodermal (Xmenf, Slug, and Snail) and endodermal (Endodermin and Sox17beta) differentiation. Given the evolutionary strategy of using common regulatory networks, it seems likely that a homologous Sox-Axis is active during embryonic development in many metazoans.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Molecular, Cellular and Developmental Biology University of Colorado at Boulder Boulder, CO 80309-0347, USA
| | | |
Collapse
|
31
|
Smith J, Wardle F, Loose M, Stanley E, Patient R. Germ layer induction in ESC--following the vertebrate roadmap. CURRENT PROTOCOLS IN STEM CELL BIOLOGY 2007; Chapter 1:Unit 1D.1. [PMID: 18785165 DOI: 10.1002/9780470151808.sc01d01s1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Controlled differentiation of pluripotential cells takes place routinely and with great success in developing vertebrate embryos. It therefore makes sense to take note of how this is achieved and use this knowledge to control the differentiation of embryonic stem cells (ESCs). An added advantage is that the differentiated cells resulting from this process in embryos have proven functionality and longevity. This unit reviews what is known about the embryonic signals that drive differentiation in one of the most informative of the vertebrate animal models of development, the amphibian Xenopus laevis. It summarizes their identities and the extent to which their activities are dose-dependent. The unit details what is known about the transcription factor responses to these signals, describing the networks of interactions that they generate. It then discusses the target genes of these transcription factors, the effectors of the differentiated state. Finally, how these same developmental programs operate during germ layer formation in the context of ESC differentiation is summarized.
Collapse
Affiliation(s)
- Jim Smith
- Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, United Kingdom
| | | | | | | | | |
Collapse
|
32
|
Saka Y, Smith JC. A mechanism for the sharp transition of morphogen gradient interpretation in Xenopus. BMC DEVELOPMENTAL BIOLOGY 2007; 7:47. [PMID: 17506890 PMCID: PMC1885807 DOI: 10.1186/1471-213x-7-47] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2007] [Accepted: 05/16/2007] [Indexed: 12/13/2022]
Abstract
Background One way in which positional information is established during embryonic development is through the graded distribution of diffusible morphogens. Unfortunately, little is known about how cells interpret different concentrations of morphogen to activate different genes or how thresholds are generated in a morphogen gradient. Results Here we show that the concentration-dependent induction of the T-box transcription factor Brachyury (Xbra) and the homeobox-containing gene Goosecoid (Gsc) by activin in Xenopus can be explained by the dynamics of a simple network consisting of three elements with a mutual negative feedback motif that can function to convert a graded signal (activin) into a binary output (Xbra on and Gsc off, or vice versa). Importantly, such a system can display sharp thresholds. Consistent with the predictions of our model, Xenopus ectodermal cells display a binary response at the single cell level after treatment with activin. Conclusion This kind of simple network with mutual negative feedback might provide a general mechanism for selective gene activation in response to different levels of a single external signal. It provides a mechanism by which a sharp boundary might be created between domains of different cell types in response to a morphogen gradient.
Collapse
Affiliation(s)
- Yasushi Saka
- Wellcome Trust/Cancer Research UK Gurdon Institute and Department of Zoology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
- Interdisciplinary Research Institute, Institut de Biologie de Lille, 1 rue du Professeur Calmette, BP447, 59021 Lille Cedex, France
| | - James C Smith
- Wellcome Trust/Cancer Research UK Gurdon Institute and Department of Zoology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| |
Collapse
|
33
|
Zorn AM, Wells JM. Molecular basis of vertebrate endoderm development. INTERNATIONAL REVIEW OF CYTOLOGY 2007; 259:49-111. [PMID: 17425939 DOI: 10.1016/s0074-7696(06)59002-3] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The embryonic endoderm gives rise to the epithelial lining of the digestive and respiratory systems and organs such as the thyroid, lungs, liver, gallbladder, and pancreas. Studies in Xenopus, zebrafish, and mice have revealed a conserved molecular pathway controlling vertebrate endoderm development. The TGFbeta/Nodal signaling pathway is at the top of this molecular hierarchy and controls the expression of a number of key transcription factors including Mix-like homeodomain proteins, Gata zinc finger factors, Sox HMG domain proteins, and Fox forkhead factors. Here we review the function of these molecules comparing and contrasting their roles in each model organism. Finally, we will describe how our understanding of the molecular pathway governing endoderm development in embryos is being used to differentiate embryonic stem cells in vitro along endodermal lineages, with the ultimate goal of making therapeutically useful tissue.
Collapse
Affiliation(s)
- Aaron M Zorn
- Division of Developmental Biology, Cincinnati Children's Hospital Research, Foundation and University of Cincinnati College of Medicine, Cincinnati, Ohio 45229, USA
| | | |
Collapse
|
34
|
Abstract
The induction of mesoderm and/or endoderm from prospective ectoderm and dorsalization of the marginal zone mesoderm may be linked to inhibition of cell cycling and DNA synthesis in early amphibian embryos. In turn, this may lead to reduction of somatic H1 histone accumulation. A greater number of cell cycles and rounds of DNA synthesis characterizes the induction of neural tissue. This is correlated with an increase of somatic H1 histone accumulation. The number of rounds of DNA replication may regulate the level of H1 histone accumulation and this may have a role in germ layer determination.
Collapse
Affiliation(s)
- Reed A Flickinger
- Department of Biological Sciences, State University of New York, Buffalo, New York 14260, USA.
| |
Collapse
|
35
|
Fletcher G, Jones GE, Patient R, Snape A. A role for GATA factors in Xenopus gastrulation movements. Mech Dev 2006; 123:730-45. [PMID: 16949798 DOI: 10.1016/j.mod.2006.07.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2006] [Revised: 07/12/2006] [Accepted: 07/16/2006] [Indexed: 01/12/2023]
Abstract
Gastrulation movements in Xenopus laevis are becoming increasingly well characterised, however the molecular mechanisms involved are less clear. Active migration of the leading edge mesendoderm across the fibronectin-coated blastocoel roof is necessary for further development of tissues such as head mesoderm, heart, blood and liver. The zinc finger transcription factors GATA4 and GATA6 are expressed in this migratory tissue during gastrulation, but their role here is unknown. This study further characterises the expression of GATA4 and 6 during gastrulation, and investigates their function in migratory behaviour. Gain-of-function experiments with these GATA factors induce cell spreading, polarisation and migration in non-motile presumptive ectoderm cells. Expression of a dominant-interfering form of GATA6, which inhibits transactivation of GATA targets, severely impairs the ability of dorsal leading edge mesendoderm to spread and translocate on fibronectin. Mosaic inhibition of GATA activity indicates that GATA factors function cell autonomously to induce cell spreading and movement in dorsal mesendoderm. Knockdown of specific GATA factors using anti-sense morpholinos indicates that GATA4 and GATA6 both contribute to dorsal mesendoderm migration in vitro. GATA4 and GATA6 are known to be involved in cell-specification of mesoderm and endoderm-derived tissues, but this is the first description of an additional role for these factors in cell migration.
Collapse
Affiliation(s)
- Georgina Fletcher
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guys Campus, London SE1 1UL, UK
| | | | | | | |
Collapse
|
36
|
Abstract
Vertebrate mesoderm induction is one of the classical problems in developmental biology. Various developmental biology approaches, particularly in Xenopus and zebrafish, have identified many of the key factors that are involved in this process and have provided major insights into how these factors interact as part of a signalling and transcription-factor network. These data are beginning to be refined by high-throughput approaches such as microarray assays. Future challenges include understanding how the prospective mesodermal cells integrate the various signals they receive and how they resolve this information to regulate their morphogenetic behaviours and cell-fate decisions.
Collapse
Affiliation(s)
- David Kimelman
- Department of Biochemistry, Box 357350, University of Washington, Seattle, Washington 98195-7350, USA.
| |
Collapse
|
37
|
Sindelka R, Ferjentsik Z, Jonák J. Developmental expression profiles of Xenopus laevis reference genes. Dev Dyn 2006; 235:754-8. [PMID: 16397894 DOI: 10.1002/dvdy.20665] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cell differentiation depends mainly on specific mRNA expression. To quantify the expression of a particular gene, the normalisation with respect to the expression of a reference gene is carried out. This is based on the assumption that the expression of the reference gene is constant during development, in different cells or tissues or after treatment. Xenopus laevis studies have frequently used eEF-1 alpha, GAPDH, ODC, L8, and H4 as reference genes. The aim of this work was to examine, by real-time RT-PCR, the expression profiles of the above-mentioned five reference genes during early development of X. laevis. It is shown that their expression profiles vary greatly during X. laevis development. The developmental changes of mRNA expression can thus significantly compromise the relative mRNA quantification based on these reference genes, when different developmental stages are to be compared. The normalisation against total RNA is recommended instead.
Collapse
Affiliation(s)
- Radek Sindelka
- Department of Gene Expression, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | | | | |
Collapse
|
38
|
Tamada H, Van Thuan N, Reed P, Nelson D, Katoku-Kikyo N, Wudel J, Wakayama T, Kikyo N. Chromatin decondensation and nuclear reprogramming by nucleoplasmin. Mol Cell Biol 2006; 26:1259-71. [PMID: 16449640 PMCID: PMC1367201 DOI: 10.1128/mcb.26.4.1259-1271.2006] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Somatic cell nuclear cloning has repeatedly demonstrated striking reversibility of epigenetic regulation of cell differentiation. Upon injection into eggs, the donor nuclei exhibit global chromatin decondensation, which might contribute to reprogramming the nuclei by derepressing dormant genes. Decondensation of sperm chromatin in eggs is explained by the replacement of sperm-specific histone variants with egg-type histones by the egg protein nucleoplasmin (Npm). However, little is known about the mechanisms of chromatin decondensation in somatic nuclei that do not contain condensation-specific histone variants. Here we found that Npm could widely decondense chromatin in undifferentiated mouse cells without overt histone exchanges but with specific epigenetic modifications that are relevant to open chromatin structure. These modifications included nucleus-wide multiple histone H3 phosphorylation, acetylation of Lys 14 in histone H3, and release of heterochromatin proteins HP1beta and TIF1beta from the nuclei. The protein kinase inhibitor staurosporine inhibited chromatin decondensation and these epigenetic modifications with the exception of H3 acetylation, potentially linking these chromatin events. At the functional level, Npm pretreatment of mouse nuclei facilitated activation of four oocyte-specific genes from the nuclei injected into Xenopus laevis oocytes. Future molecular elucidation of chromatin decondensation by Npm will significantly contribute to our understanding of the plasticity of cell differentiation.
Collapse
Affiliation(s)
- Hiroshi Tamada
- Stem Cell Institute, Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, MMC 716, 420 Delaware St. SE, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
This Teaching Resource provides a summary and slides derived from a lecture on the signaling cascades that mediate early embryogenesis and is part of the course "Cell Signaling Systems: A Course for Graduate Students." The lecture focuses on the mechanisms underlying germ layer formation, patterning, and suppression in the vertebrate embryo, with particular emphasis on the regulation of mesodermal development in the model organism Xenopus laevis.
Collapse
Affiliation(s)
- Daniel C Weinstein
- Department of Pharmacology and Biological Chemistry, Mount Sinai School of Medicine, New York, NY 10029, USA.
| |
Collapse
|
40
|
Kaern M, Elston TC, Blake WJ, Collins JJ. Stochasticity in gene expression: from theories to phenotypes. Nat Rev Genet 2005; 6:451-64. [PMID: 15883588 DOI: 10.1038/nrg1615] [Citation(s) in RCA: 1537] [Impact Index Per Article: 76.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Genetically identical cells exposed to the same environmental conditions can show significant variation in molecular content and marked differences in phenotypic characteristics. This variability is linked to stochasticity in gene expression, which is generally viewed as having detrimental effects on cellular function with potential implications for disease. However, stochasticity in gene expression can also be advantageous. It can provide the flexibility needed by cells to adapt to fluctuating environments or respond to sudden stresses, and a mechanism by which population heterogeneity can be established during cellular differentiation and development.
Collapse
Affiliation(s)
- Mads Kaern
- Department of Cellular and Molecular Medicine and Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H8M5, Canada.
| | | | | | | |
Collapse
|
41
|
Suri C, Haremaki T, Weinstein DC. Xema, a foxi-class gene expressed in the gastrula stage Xenopus ectoderm, is required for the suppression of mesendoderm. Development 2005; 132:2733-42. [PMID: 15901660 PMCID: PMC3525708 DOI: 10.1242/dev.01865] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The molecular basis of vertebrate germ layer formation has been the focus of intense scrutiny for decades, and the inductive interactions underlying this process are well defined. Only recently, however, have studies demonstrated that the regulated inhibition of ectopic germ layer formation is also crucial for patterning the early vertebrate embryo. We report here the characterization of Xema (Xenopus Ectodermally-expressed Mesendoderm Antagonist), a novel member of the Foxi-subclass of winged-helix transcription factors that is involved in the suppression of ectopic germ layer formation in the frog, Xenopus laevis. Xema transcripts are restricted to the animal pole ectoderm during early Xenopus development. Ectopic expression of Xema RNA inhibits mesoderm induction, both by growth factors and in the marginal zone, in vivo. Conversely, introduction of antisense morpholino oligonucleotides directed against the Xema transcript stimulates the expression of a broad range of mesodermal and endodermal marker genes in the animal pole. Our studies demonstrate that Xema is both necessary and sufficient for the inhibition of ectopic mesendoderm in the cells of the presumptive ectoderm, and support a model in which Fox proteins function in part to restrict inappropriate germ layer development throughout the vertebrate embryo.
Collapse
|
42
|
Messenger NJ, Kabitschke C, Andrews R, Grimmer D, Núñez Miguel R, Blundell TL, Smith JC, Wardle FC. Functional Specificity of the Xenopus T-Domain Protein Brachyury Is Conferred by Its Ability to Interact with Smad1. Dev Cell 2005; 8:599-610. [PMID: 15809041 DOI: 10.1016/j.devcel.2005.03.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2004] [Revised: 11/01/2004] [Accepted: 03/04/2005] [Indexed: 11/30/2022]
Abstract
Members of the T-box gene family play important and diverse roles in development and disease. Here, we study the functional specificities of the Xenopus T-domain proteins Xbra and VegT, which differ in their abilities to induce gene expression in prospective ectodermal tissue. In particular, VegT induces strong expression of goosecoid whereas Xbra cannot. Our results indicate that Xbra is unable to induce goosecoid because it directly activates expression of Xom, a repressor of goosecoid that acts downstream of BMP signaling. We show that the inability of Xbra to induce goosecoid is imposed by an N-terminal domain that interacts with the C-terminal MH2 domain of Smad1, a component of the BMP signal transduction pathway. Interference with this interaction causes ectopic activation of goosecoid and anteriorization of the embryo. These findings suggest a mechanism by which individual T-domain proteins may interact with different partners to elicit a specific response.
Collapse
Affiliation(s)
- Nigel J Messenger
- Wellcome Trust/Cancer Research UK Gurdon Institute, Department of Zoology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Cinquin O, Demongeot J. High-dimensional switches and the modelling of cellular differentiation. J Theor Biol 2004; 233:391-411. [PMID: 15652148 DOI: 10.1016/j.jtbi.2004.10.027] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2004] [Revised: 10/13/2004] [Accepted: 10/14/2004] [Indexed: 12/31/2022]
Abstract
Many genes have been identified as driving cellular differentiation, but because of their complex interactions, the understanding of their collective behaviour requires mathematical modelling. Intriguingly, it has been observed in numerous developmental contexts, and particularly haematopoiesis, that genes regulating differentiation are initially co-expressed in progenitors despite their antagonism, before one is upregulated and others downregulated. We characterise conditions under which three classes of generic "master regulatory networks", modelled at the molecular level after experimentally observed interactions (including bHLH protein dimerisation), and including an arbitrary number of antagonistic components, can behave as a "multi-switch", directing differentiation in an all-or-none fashion to a specific cell-type chosen among more than two possible outcomes. bHLH dimerisation networks can readily display coexistence of many antagonistic factors when competition is low (a simple characterisation is derived). Decision-making can be forced by a transient increase in competition, which could correspond to some unexplained experimental observations related to Id proteins; the speed of response varies with the initial conditions the network is subjected to, which could explain some aspects of cell behaviour upon reprogramming. The coexistence of antagonistic factors at low levels, early in the differentiation process or in pluripotent stem cells, could be an intrinsic property of the interaction between those factors, not requiring a specific regulatory system.
Collapse
Affiliation(s)
- Olivier Cinquin
- CoMPLEX, University College London, Gower Street, London WC1E 6BT, UK.
| | | |
Collapse
|