1
|
Strauss ME, Ton MLN, Mason S, Bagri J, Harland LT, Imaz-Rosshandler I, Wilson NK, Nichols J, Tyser RC, Göttgens B, Marioni JC, Guibentif C. A single-cell and tissue-scale analysis suite resolves Mixl1's role in heart development. iScience 2025; 28:112397. [PMID: 40330894 PMCID: PMC12051648 DOI: 10.1016/j.isci.2025.112397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 12/10/2024] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Perturbation studies using gene knockouts have become a key tool for understanding the roles of regulatory genes in development. However, large-scale studies dissecting the molecular role of development master regulators in every cell type throughout the embryo are technically challenging and scarce. Here, we systematically characterize the knockout effects of the key developmental regulators T/Brachyury and Mixl1 in gastrulation and early organogenesis using single-cell profiling of chimeric mouse embryos. For the analysis of these experimental data, we present COSICC, an effective suite of statistical tools to characterize perturbation effects in complex developing cell populations. We gain insights into T's role in lateral plate mesoderm, limb development, and posterior intermediate mesoderm specification. Furthermore, we generate Mixl1 -/- embryonic chimeras and reveal the role of this key transcription factor in discrete mesoderm lineages, in particular concerning developmental dysregulation of the recently identified juxta-cardiac field.
Collapse
Affiliation(s)
- Magdalena E. Strauss
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, Cambridge CB10 1SD, UK
- Department of Mathematics and Statistics, University of Exeter, Exeter EX4 4PY, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Mai-Linh Nu Ton
- Department of Haematology, University of Cambridge, Cambridge CB2 0AW, UK
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
| | - Samantha Mason
- Department of Haematology, University of Cambridge, Cambridge CB2 0AW, UK
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
| | - Jaana Bagri
- Department of Haematology, University of Cambridge, Cambridge CB2 0AW, UK
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
| | - Luke T.G. Harland
- Department of Haematology, University of Cambridge, Cambridge CB2 0AW, UK
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
| | | | - Nicola K. Wilson
- Department of Haematology, University of Cambridge, Cambridge CB2 0AW, UK
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
| | - Jennifer Nichols
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Richard C.V. Tyser
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
| | - Berthold Göttgens
- Department of Haematology, University of Cambridge, Cambridge CB2 0AW, UK
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
| | - John C. Marioni
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, Cambridge CB10 1SD, UK
| | - Carolina Guibentif
- Institute Biomedicine, Department of Microbiology and Immunology, Sahlgrenska Center for Cancer Research, University of Gothenburg, 413 90 Gothenburg, Sweden
| |
Collapse
|
2
|
Osteil P, Withey S, Santucci N, Aryamanesh N, Pang I, Salehin N, Sun J, Qin A, Su J, Knowles H, Li XB, Cai S, Wolvetang E, Tam PPL. MIXL1 activation in endoderm differentiation of human induced pluripotent stem cells. Stem Cell Reports 2025; 20:102482. [PMID: 40280138 DOI: 10.1016/j.stemcr.2025.102482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) possess the ability to differentiate into a multitude of cell and tissue types but display heterogeneous propensity of differentiation into specific lineage. Characterization of the transcriptome of 11 hiPSC lines showed that activation of MIXL1 at the early stage of stem cell differentiation correlated with higher efficacy in generating definitive endoderm and advancing differentiation and maturation of endoderm derivatives. Enforced expression of MIXL1 in the endoderm-inefficient hiPSCs enhanced the propensity of endoderm differentiation, suggesting that modulation of key drivers of lineage differentiation can re-wire hiPSC to the desired lineage propensity to generate the requisite stem cell products.
Collapse
Affiliation(s)
- Pierre Osteil
- Embryology Research Unit, Children's Medical Research Institute, University of Sydney, Sydney, Australia.
| | - Sarah Withey
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Australia
| | - Nicole Santucci
- Embryology Research Unit, Children's Medical Research Institute, University of Sydney, Sydney, Australia
| | - Nader Aryamanesh
- Bioinformatics Group, Children's Medical Research Institute, University of Sydney, Sydney, Australia
| | - Ignatius Pang
- Bioinformatics Group, Children's Medical Research Institute, University of Sydney, Sydney, Australia
| | - Nazmus Salehin
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Jane Sun
- Embryology Research Unit, Children's Medical Research Institute, University of Sydney, Sydney, Australia
| | - Annie Qin
- Embryology Research Unit, Children's Medical Research Institute, University of Sydney, Sydney, Australia
| | - Jiayi Su
- Embryology Research Unit, Children's Medical Research Institute, University of Sydney, Sydney, Australia
| | - Hilary Knowles
- Embryology Research Unit, Children's Medical Research Institute, University of Sydney, Sydney, Australia
| | - Xiucheng Bella Li
- Embryology Research Unit, Children's Medical Research Institute, University of Sydney, Sydney, Australia
| | - Simon Cai
- Bioinformatics Group, Children's Medical Research Institute, University of Sydney, Sydney, Australia
| | - Ernst Wolvetang
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Australia
| | - Patrick P L Tam
- Embryology Research Unit, Children's Medical Research Institute, University of Sydney, Sydney, Australia; School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, Australia.
| |
Collapse
|
3
|
Masamsetti VP, Salehin N, Kim HJ, Santucci N, Weatherstone M, McMahon R, Marshall LL, Knowles H, Sun J, Studdert JB, Aryamanesh N, Wang R, Jing N, Yang P, Osteil P, Tam PPL. Lineage contribution of the mesendoderm progenitors in the gastrulating mouse embryo. Dev Cell 2025:S1534-5807(25)00120-0. [PMID: 40132585 DOI: 10.1016/j.devcel.2025.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/08/2024] [Accepted: 02/28/2025] [Indexed: 03/27/2025]
Abstract
A population of putative mesendoderm progenitors that can contribute cellular descendants to both mesoderm and endoderm lineages is identified in the gastrulating mouse embryo. These progenitor cells are localized to the posterior epiblast, primitive streak, and nascent mesoderm of mid-streak- (E7.0) to late-streak-stage (E7.5) embryos. Lineage tracing in vivo identified that putative mesendoderm progenitors contribute descendants to the definitive endoderm and the axial mesendoderm of E7.75 embryos and to the endoderm of the foregut and hindgut of the E8.5-8.75 embryos. Differentiation of mouse epiblast stem cells identified that the choice between endoderm and mesoderm cell fates depends on the timing of Mixl1 activation upon exit from pluripotency. The knowledge gained on the spatiotemporal distribution of mesendoderm progenitors and the molecular drivers underpinning the divergence of cell lineages in these progenitors enriches our mechanistic understanding of the allocation of the tissue progenitors to germ layer derivatives in early development.
Collapse
Affiliation(s)
- V Pragathi Masamsetti
- Embryology Research Unit, Children's Medical Research Institute, Westmead, NSW, Australia; School of Medical Science, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia.
| | - Nazmus Salehin
- Embryology Research Unit, Children's Medical Research Institute, Westmead, NSW, Australia; School of Medical Science, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Hani Jieun Kim
- Computational Systems Biology Unit, Children's Medical Research Institute, Westmead, NSW, Australia; School of Mathematics and Statistics, University of Sydney, Sydney, NSW, Australia
| | - Nicole Santucci
- Embryology Research Unit, Children's Medical Research Institute, Westmead, NSW, Australia
| | - Megan Weatherstone
- Embryology Research Unit, Children's Medical Research Institute, Westmead, NSW, Australia
| | - Riley McMahon
- Embryology Research Unit, Children's Medical Research Institute, Westmead, NSW, Australia; School of Medical Science, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Lee L Marshall
- Bioinformatics Group, Children's Medical Research Institute, Westmead, NSW, Australia
| | - Hilary Knowles
- Embryology Research Unit, Children's Medical Research Institute, Westmead, NSW, Australia
| | - Jane Sun
- Embryology Research Unit, Children's Medical Research Institute, Westmead, NSW, Australia
| | - Josh B Studdert
- Embryology Research Unit, Children's Medical Research Institute, Westmead, NSW, Australia
| | - Nader Aryamanesh
- Embryology Research Unit, Children's Medical Research Institute, Westmead, NSW, Australia; School of Medical Science, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Bioinformatics Group, Children's Medical Research Institute, Westmead, NSW, Australia
| | - Ran Wang
- Biomedical Informatics & Genomics Center, Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China
| | - Naihe Jing
- Guangzhou National Laboratory, Guangzhou 510005, China
| | - Pengyi Yang
- Computational Systems Biology Unit, Children's Medical Research Institute, Westmead, NSW, Australia; School of Mathematics and Statistics, University of Sydney, Sydney, NSW, Australia
| | - Pierre Osteil
- Embryology Research Unit, Children's Medical Research Institute, Westmead, NSW, Australia
| | - Patrick P L Tam
- Embryology Research Unit, Children's Medical Research Institute, Westmead, NSW, Australia; School of Medical Science, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
4
|
Schröder CM, Zissel L, Mersiowsky SL, Tekman M, Probst S, Schüle KM, Preissl S, Schilling O, Timmers HTM, Arnold SJ. EOMES establishes mesoderm and endoderm differentiation potential through SWI/SNF-mediated global enhancer remodeling. Dev Cell 2025; 60:735-748.e5. [PMID: 39662466 DOI: 10.1016/j.devcel.2024.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 09/17/2024] [Accepted: 11/14/2024] [Indexed: 12/13/2024]
Abstract
Mammalian pluripotent cells first segregate into neuroectoderm (NE), or mesoderm and endoderm (ME), characterized by lineage-specific transcriptional programs and chromatin states. To date, the relationship between transcription factor activities and dynamic chromatin changes that guide cell specification remains ill-defined. In this study, we employ mouse embryonic stem cell differentiation toward ME lineages to reveal crucial roles of the Tbx factor Eomes to globally establish ME enhancer accessibility as the prerequisite for ME lineage competence and ME-specific gene expression. EOMES cooperates with the SWItch/sucrose non-fermentable (SWI/SNF) complex to drive chromatin rewiring that is essential to overcome default NE differentiation, which is favored by asymmetries in chromatin accessibility at pluripotent state. Following global ME enhancer remodeling, ME-specific gene transcription is controlled by additional signals such as Wnt and transforming growth factor β (TGF-β)/NODAL, as a second layer of gene expression regulation, which can be mechanistically separated from initial chromatin remodeling activities.
Collapse
Affiliation(s)
- Chiara M Schröder
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany; CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
| | - Lea Zissel
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Sophie-Luise Mersiowsky
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Mehmet Tekman
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Simone Probst
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Katrin M Schüle
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
| | - Sebastian Preissl
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
| | - Oliver Schilling
- Institute for Surgical Pathology, Medical Centre, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - H Th Marc Timmers
- Department of Urology, Medical Centre, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; German Cancer Consortium (DKTK), partner site Freiburg, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sebastian J Arnold
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg, Germany; CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany.
| |
Collapse
|
5
|
Azbazdar Y, De Robertis EM. Double assurance in the induction of axial development by egg dorsal determinants in Xenopus embryos. Proc Natl Acad Sci U S A 2025; 122:e2421772122. [PMID: 39928870 PMCID: PMC11848351 DOI: 10.1073/pnas.2421772122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/04/2025] [Indexed: 02/12/2025] Open
Abstract
We recently reported that microinjection of Xenopus nodal-related (xnr) mRNAs into β-catenin-depleted Xenopus embryos rescued a complete dorsal axis. Xnrs mediate the signal of the Nieuwkoop center that induces the Spemann-Mangold organizer in the overlying mesoderm, a process inhibited by the Nodal antagonist Cerberus-short (CerS). However, β-catenin also induces a second signaling center in the dorsal prospective ectoderm, designated the Blastula Chordin and Noggin Expression (BCNE) center, in which the homeobox gene siamois (sia) plays a major role. In this study, we asked whether the Xnrs and Sia depend on each other or function on parallel pathways. Expression of both genes induced β-catenin-depleted embryos to form complete axes with heads and eyes via the activation of similar sets of downstream organizer-specific genes. Xnrs did not activate siamois, and, conversely, Sia did not activate xnrs, although both were induced by β-catenin stabilization. Depletion with morpholinos revealed a robust role for the downstream target Chordin. Remarkably, Chordin depletion prevented all ectopic effects resulting from microinjection of the mRNA encoding the maternal cytoplasmic determinant Huluwa, including the radial expansion of brain tissue and the ectopic expression of the ventral gene sizzled. The main conclusion was that the BCNE and Nieuwkoop centers provide a double assurance mechanism for axial formation by independently activating similar downstream transcriptional target gene repertoires. We suggest that Siamois likely evolved from an ancestral Mix-type homeodomain protein called Sebox as a Xenopus-specific adaptation for the rapid differentiation of the anterior neural plate in the ectoderm.
Collapse
Affiliation(s)
- Yagmur Azbazdar
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA90095-1662
| | - Edward M. De Robertis
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA90095-1662
| |
Collapse
|
6
|
Tam PPL, Masamsetti P. Functional attributes of the anterior mesendoderm in patterning the anterior neural structures during head formation in the mouse. Cells Dev 2025:203999. [PMID: 39880304 DOI: 10.1016/j.cdev.2025.203999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 01/31/2025]
Abstract
Induction of the neural ectoderm and the patterning of embryonic brain are the requisite organizing activity for head formation. Studies of loss-of-function mouse mutants that displayed a head truncation phenotype pointed to a key functional role of the anterior mesendoderm in anterior neural patterning. In this overview, we highlight the learning of the molecular attributes underpinning the formation of the anterior mesendoderm, the acquisition of ectoderm competence in the epiblast and the patterning of the embryonic brain during gastrulation and neurulation.
Collapse
Affiliation(s)
- Patrick P L Tam
- Embryology Research Unit, Children's Medical Research Institute, University of Sydney, and School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, NSW, Australia.
| | - Pragathi Masamsetti
- Embryology Research Unit, Children's Medical Research Institute, University of Sydney, and School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, NSW, Australia
| |
Collapse
|
7
|
Robles-Garcia M, Thimonier C, Angoura K, Ozga E, MacPherson H, Blin G. In vitro modelling of anterior primitive streak patterning with human pluripotent stem cells identifies the path to notochord progenitors. Development 2024; 151:dev202983. [PMID: 39611739 DOI: 10.1242/dev.202983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 11/20/2024] [Indexed: 11/30/2024]
Abstract
Notochord progenitors (NotoPs) represent a scarce yet crucial embryonic cell population, playing important roles in embryo patterning and eventually giving rise to the cells that form and maintain intervertebral discs. The mechanisms regulating NotoPs emergence are unclear. This knowledge gap persists due to the inherent complexity of cell fate patterning during gastrulation, particularly within the anterior primitive streak (APS), where NotoPs first arise alongside neuro-mesoderm and endoderm. To gain insights into this process, we use micropatterning together with FGF and the WNT pathway activator CHIR9901 to guide the development of human embryonic stem cells into reproducible patterns of APS cell fates. We show that CHIR9901 dosage dictates the downstream dynamics of endogenous TGFβ signalling, which in turn controls cell fate decisions. While sustained NODAL signalling defines endoderm and NODAL inhibition is imperative for neuro-mesoderm emergence, timely inhibition of NODAL signalling with spatial confinement potentiates WNT activity and enables us to generate NotoPs efficiently. Our work elucidates the signalling regimes underpinning NotoP emergence and provides insights into the regulatory mechanisms controlling the balance of APS cell fates during gastrulation.
Collapse
Affiliation(s)
- Miguel Robles-Garcia
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, EH16 4UU, UK
- Institute for Stem Cell Research, School of Biological Sciences, The University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Chloë Thimonier
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, EH16 4UU, UK
- Institute for Stem Cell Research, School of Biological Sciences, The University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Konstantina Angoura
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, EH16 4UU, UK
- Institute for Stem Cell Research, School of Biological Sciences, The University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Ewa Ozga
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, EH16 4UU, UK
- Institute for Stem Cell Research, School of Biological Sciences, The University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Heather MacPherson
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, EH16 4UU, UK
- Institute for Stem Cell Research, School of Biological Sciences, The University of Edinburgh, Edinburgh, EH16 4UU, UK
| | - Guillaume Blin
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, EH16 4UU, UK
- Institute for Stem Cell Research, School of Biological Sciences, The University of Edinburgh, Edinburgh, EH16 4UU, UK
| |
Collapse
|
8
|
Rüegg AB, van der Weijden VA, de Sousa JA, von Meyenn F, Pausch H, Ulbrich SE. Developmental progression continues during embryonic diapause in the roe deer. Commun Biol 2024; 7:270. [PMID: 38443549 PMCID: PMC10914810 DOI: 10.1038/s42003-024-05944-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/21/2024] [Indexed: 03/07/2024] Open
Abstract
Embryonic diapause in mammals is a temporary developmental delay occurring at the blastocyst stage. In contrast to other diapausing species displaying a full arrest, the blastocyst of the European roe deer (Capreolus capreolus) proliferates continuously and displays considerable morphological changes in the inner cell mass. We hypothesised that developmental progression also continues during this period. Here we evaluate the mRNA abundance of developmental marker genes in embryos during diapause and elongation. Our results show that morphological rearrangements of the epiblast during diapause correlate with gene expression patterns and changes in cell polarity. Immunohistochemical staining further supports these findings. Primitive endoderm formation occurs during diapause in embryos composed of around 3,000 cells. Gastrulation coincides with elongation and thus takes place after embryo reactivation. The slow developmental progression makes the roe deer an interesting model for unravelling the link between proliferation and differentiation and requirements for embryo survival.
Collapse
Affiliation(s)
- Anna B Rüegg
- ETH Zurich, Animal Physiology, Institute of Agricultural Sciences, Zurich, Switzerland
| | - Vera A van der Weijden
- ETH Zurich, Animal Physiology, Institute of Agricultural Sciences, Zurich, Switzerland
- Max-Planck Institute for Molecular Genetics, Berlin, Germany
| | - João Agostinho de Sousa
- ETH Zurich, Laboratory of Nutrition and Metabolic Epigenetics, Institute of Food, Nutrition and Health, Zurich, Switzerland
| | - Ferdinand von Meyenn
- ETH Zurich, Laboratory of Nutrition and Metabolic Epigenetics, Institute of Food, Nutrition and Health, Zurich, Switzerland
| | - Hubert Pausch
- ETH Zurich, Animal Genomics, Institute of Agricultural Sciences, Zurich, Switzerland
| | - Susanne E Ulbrich
- ETH Zurich, Animal Physiology, Institute of Agricultural Sciences, Zurich, Switzerland.
| |
Collapse
|
9
|
Lin Y, Wu TY, Chen X, Wan S, Chao B, Xin J, Yang JYH, Wong WH, Wang YXR. Data integration and inference of gene regulation using single-cell temporal multimodal data with scTIE. Genome Res 2024; 34:119-133. [PMID: 38190633 PMCID: PMC10903952 DOI: 10.1101/gr.277960.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 12/13/2023] [Indexed: 01/10/2024]
Abstract
Single-cell technologies offer unprecedented opportunities to dissect gene regulatory mechanisms in context-specific ways. Although there are computational methods for extracting gene regulatory relationships from scRNA-seq and scATAC-seq data, the data integration problem, essential for accurate cell type identification, has been mostly treated as a standalone challenge. Here we present scTIE, a unified method that integrates temporal multimodal data and infers regulatory relationships predictive of cellular state changes. scTIE uses an autoencoder to embed cells from all time points into a common space by using iterative optimal transport, followed by extracting interpretable information to predict cell trajectories. Using a variety of synthetic and real temporal multimodal data sets, we show scTIE achieves effective data integration while preserving more biological signals than existing methods, particularly in the presence of batch effects and noise. Furthermore, on the exemplar multiome data set we generated from differentiating mouse embryonic stem cells over time, we show scTIE captures regulatory elements highly predictive of cell transition probabilities, providing new potentials to understand the regulatory landscape driving developmental processes.
Collapse
Affiliation(s)
- Yingxin Lin
- School of Mathematics and Statistics, The University of Sydney, NSW 2006, Australia
- Charles Perkins Centre, The University of Sydney, NSW 2006, Australia
- Laboratory of Data Discovery for Health Limited (D24H), Science Park, Hong Kong SAR 999077, China
| | - Tung-Yu Wu
- Department of Statistics, Stanford University, Stanford, California 94305-4020, USA
| | - Xi Chen
- Department of Statistics, Stanford University, Stanford, California 94305-4020, USA
| | - Sheng Wan
- Institute of Electronics, National Yang Ming Chiao Tung University, Hsinchu 30010, Taiwan
| | - Brian Chao
- Department of Electrical Engineering, Stanford University, Stanford, California 94305-9505, USA
| | - Jingxue Xin
- Department of Statistics, Stanford University, Stanford, California 94305-4020, USA
| | - Jean Y H Yang
- School of Mathematics and Statistics, The University of Sydney, NSW 2006, Australia
- Charles Perkins Centre, The University of Sydney, NSW 2006, Australia
- Laboratory of Data Discovery for Health Limited (D24H), Science Park, Hong Kong SAR 999077, China
| | - Wing H Wong
- Department of Statistics, Stanford University, Stanford, California 94305-4020, USA;
- Department of Biomedical Data Science, Stanford University, Stanford, California 94305-5464, USA
- Bio-X Program, Stanford University, Stanford, California 94305, USA
| | - Y X Rachel Wang
- School of Mathematics and Statistics, The University of Sydney, NSW 2006, Australia;
| |
Collapse
|
10
|
Ding S, Zhang R, Zhang P, Shi J, Liu L, Li J, Zhang R, Wu F, Zhou P. The application of quantitative telomerase activity measurement as an important indicator to monitor the cardiomyocyte differentiation process of human induced pluripotent stem cells under defined conditions. Biochem Biophys Res Commun 2023; 687:149150. [PMID: 37939503 DOI: 10.1016/j.bbrc.2023.149150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/11/2023] [Accepted: 10/23/2023] [Indexed: 11/10/2023]
Abstract
The construction of an in vitro differentiation system for human induced pluripotent stem cells (hiPSCs) has made exciting progress, but it is still of great significance to clarify the differentiation process. The use of conventional genetic and protein-labeled microscopes to observe or detect different stages of hiPSC differentiation is not specific enough and is cumbersome and time-consuming. In this study, in addition to analyzing the expression of gene/protein-related markers, we used a previously reported simple and excellent quantitative method of cellular telomerase activity based on a quartz crystal microbalance (TREAQ) device to monitor the dynamic changes in cellular telomerase activity in hiPSCs during myocardial differentiation under chemically defined conditions. Finally, by integrating these results, we analyzed the relationship between telomerase activity and the expression of marker genes/proteins as well as the cell type at each study time point. This dynamic quantitative measurement of cellular telomerase activity should be a promising indicator for monitoring dynamic changes in a stage of hiPSC differentiation and inducing cell types. This study provided a quantitative, dynamic and simple monitoring index for the in vitro differentiation process of hiPSC-CMs, which was a certain reference value for the optimization and improvement of the induction system.
Collapse
Affiliation(s)
- Shaoli Ding
- Department of Pain Treatment, Gansu Provincial Hospital, Lanzhou, China; The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Rongzhi Zhang
- Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Pengxia Zhang
- School of Stomatology, Lanzhou University, No.222 Tianshui South Road, Chengguan District, Lanzhou, 730000, China
| | - Jiamin Shi
- School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Lu Liu
- Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Jiamin Li
- School of Stomatology, Lanzhou University, No.222 Tianshui South Road, Chengguan District, Lanzhou, 730000, China
| | - Rui Zhang
- School of Stomatology, Lanzhou University, No.222 Tianshui South Road, Chengguan District, Lanzhou, 730000, China
| | - Fujian Wu
- Translational Medicine Collaborative Innovation Center, Shenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, 518055, Guangdong, China.
| | - Ping Zhou
- School of Stomatology, Lanzhou University, No.222 Tianshui South Road, Chengguan District, Lanzhou, 730000, China.
| |
Collapse
|
11
|
Miura A, Sarmah H, Tanaka J, Hwang Y, Sawada A, Shimamura Y, Otoshi T, Kondo Y, Fang Y, Shimizu D, Ninish Z, Suer JL, Dubois NC, Davis J, Toyooka S, Wu J, Que J, Hawkins FJ, Lin CS, Mori M. Conditional blastocyst complementation of a defective Foxa2 lineage efficiently promotes the generation of the whole lung. eLife 2023; 12:e86105. [PMID: 37861292 PMCID: PMC10642968 DOI: 10.7554/elife.86105] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 10/19/2023] [Indexed: 10/21/2023] Open
Abstract
Millions suffer from incurable lung diseases, and the donor lung shortage hampers organ transplants. Generating the whole organ in conjunction with the thymus is a significant milestone for organ transplantation because the thymus is the central organ to educate immune cells. Using lineage-tracing mice and human pluripotent stem cell (PSC)-derived lung-directed differentiation, we revealed that gastrulating Foxa2 lineage contributed to both lung mesenchyme and epithelium formation. Interestingly, Foxa2 lineage-derived cells in the lung mesenchyme progressively increased and occupied more than half of the mesenchyme niche, including endothelial cells, during lung development. Foxa2 promoter-driven, conditional Fgfr2 gene depletion caused the lung and thymus agenesis phenotype in mice. Wild-type donor mouse PSCs injected into their blastocysts rescued this phenotype by complementing the Fgfr2-defective niche in the lung epithelium and mesenchyme and thymic epithelium. Donor cell is shown to replace the entire lung epithelial and robust mesenchymal niche during lung development, efficiently complementing the nearly entire lung niche. Importantly, those mice survived until adulthood with normal lung function. These results suggest that our Foxa2 lineage-based model is unique for the progressive mobilization of donor cells into both epithelial and mesenchymal lung niches and thymus generation, which can provide critical insights into studying lung transplantation post-transplantation shortly.
Collapse
Affiliation(s)
- Akihiro Miura
- Columbia Center for Human Development and Division of Pulmonary, Allergy, Critical Care, Department of Medicine, Columbia University Medical CenterNew YorkUnited States
- Department of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayamaJapan
| | - Hemanta Sarmah
- Columbia Center for Human Development and Division of Pulmonary, Allergy, Critical Care, Department of Medicine, Columbia University Medical CenterNew YorkUnited States
| | - Junichi Tanaka
- Columbia Center for Human Development and Division of Pulmonary, Allergy, Critical Care, Department of Medicine, Columbia University Medical CenterNew YorkUnited States
| | - Youngmin Hwang
- Columbia Center for Human Development and Division of Pulmonary, Allergy, Critical Care, Department of Medicine, Columbia University Medical CenterNew YorkUnited States
| | - Anri Sawada
- Columbia Center for Human Development and Division of Pulmonary, Allergy, Critical Care, Department of Medicine, Columbia University Medical CenterNew YorkUnited States
| | - Yuko Shimamura
- Columbia Center for Human Development and Division of Pulmonary, Allergy, Critical Care, Department of Medicine, Columbia University Medical CenterNew YorkUnited States
| | - Takehiro Otoshi
- Columbia Center for Human Development and Division of Pulmonary, Allergy, Critical Care, Department of Medicine, Columbia University Medical CenterNew YorkUnited States
| | - Yuri Kondo
- Columbia Center for Human Development and Division of Pulmonary, Allergy, Critical Care, Department of Medicine, Columbia University Medical CenterNew YorkUnited States
| | - Yinshan Fang
- Columbia Center for Human Development and Division of Pulmonary, Allergy, Critical Care, Department of Medicine, Columbia University Medical CenterNew YorkUnited States
| | - Dai Shimizu
- Columbia Center for Human Development and Division of Pulmonary, Allergy, Critical Care, Department of Medicine, Columbia University Medical CenterNew YorkUnited States
| | - Zurab Ninish
- Columbia Center for Human Development and Division of Pulmonary, Allergy, Critical Care, Department of Medicine, Columbia University Medical CenterNew YorkUnited States
| | - Jake Le Suer
- The Pulmonary Center and Department of Medicine, Boston University School of MedicineBostonUnited States
- Center for Regenerative Medicine, Boston University and Boston Medical CenterBostonUnited States
| | - Nicole C Dubois
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Jennifer Davis
- Department of Pathology, University of WashingtonSeattleUnited States
| | - Shinichi Toyooka
- Department of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayamaJapan
| | - Jun Wu
- Department of Molecular Biology, University of Texas Southwestern Medical CenterDallasUnited States
| | - Jianwen Que
- Columbia Center for Human Development and Division of Pulmonary, Allergy, Critical Care, Department of Medicine, Columbia University Medical CenterNew YorkUnited States
| | - Finn J Hawkins
- The Pulmonary Center and Department of Medicine, Boston University School of MedicineBostonUnited States
- Center for Regenerative Medicine, Boston University and Boston Medical CenterBostonUnited States
| | - Chyuan-Sheng Lin
- Bernard and Shirlee Brown Glaucoma Laboratory, Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University Irving Medical CenterNew YorkUnited States
| | - Munemasa Mori
- Columbia Center for Human Development and Division of Pulmonary, Allergy, Critical Care, Department of Medicine, Columbia University Medical CenterNew YorkUnited States
| |
Collapse
|
12
|
Glover HJ, Holliday H, Shparberg RA, Winkler D, Day M, Morris MB. Signalling pathway crosstalk stimulated by L-proline drives mouse embryonic stem cells to primitive-ectoderm-like cells. Development 2023; 150:dev201704. [PMID: 37823343 PMCID: PMC10652046 DOI: 10.1242/dev.201704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 10/02/2023] [Indexed: 10/13/2023]
Abstract
The amino acid L-proline exhibits growth factor-like properties during development - from improving blastocyst development to driving neurogenesis in vitro. Addition of 400 μM L-proline to self-renewal medium drives naïve mouse embryonic stem cells (ESCs) to early primitive ectoderm-like (EPL) cells - a transcriptionally distinct primed or partially primed pluripotent state. EPL cells retain expression of pluripotency genes, upregulate primitive ectoderm markers, undergo a morphological change and have increased cell number. These changes are facilitated by a complex signalling network hinging on the Mapk, Fgfr, Pi3k and mTor pathways. Here, we use a factorial experimental design coupled with statistical modelling to understand which signalling pathways are involved in the transition between ESCs and EPL cells, and how they underpin changes in morphology, cell number, apoptosis, proliferation and gene expression. This approach reveals pathways which work antagonistically or synergistically. Most properties were affected by more than one inhibitor, and each inhibitor blocked specific aspects of the naïve-to-primed transition. These mechanisms underpin progression of stem cells across the in vitro pluripotency continuum and serve as a model for pre-, peri- and post-implantation embryogenesis.
Collapse
Affiliation(s)
- Hannah J. Glover
- School of Medical Sciences, University of Sydney, Sydney 2006, Australia
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Holly Holliday
- School of Medical Sciences, University of Sydney, Sydney 2006, Australia
| | | | - David Winkler
- Department of Biochemistry and Chemistry, Latrobe Institute for Molecular Science, Latrobe University, Bundoora 3083, Australia
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville 3052, Australia
- Advanced Materials and Healthcare Technologies, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK
| | - Margot Day
- School of Medical Sciences, University of Sydney, Sydney 2006, Australia
| | - Michael B. Morris
- School of Medical Sciences, University of Sydney, Sydney 2006, Australia
| |
Collapse
|
13
|
Nikaido M, Shirai A, Mizumaki Y, Shigenobu S, Ueno N, Hatta K. Intestinal expression patterns of transcription factors and markers for interstitial cells in the larval zebrafish. Dev Growth Differ 2023; 65:418-428. [PMID: 37452633 DOI: 10.1111/dgd.12878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/26/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
For the digestion of food, it is important for the gut to be differentiated regionally and to have proper motor control. However, the number of transcription factors that regulate its development is still limited. Meanwhile, the interstitial cells of the gastrointestinal (GI) tract are necessary for intestinal motility in addition to the enteric nervous system. There are anoctamine1 (Ano1)-positive and platelet-derived growth factor receptor α (Pdgfra)-positive interstitial cells in mammal, but Pdgfra-positive cells have not been reported in the zebrafish. To identify new transcription factors involved in GI tract development, we used RNA sequencing comparing between larval and adult gut. We isolated 40 transcription factors that were more highly expressed in the larval gut. We demonstrated expression patterns of the 13 genes, 7 of which were newly found to be expressed in the zebrafish larval gut. Six of the 13 genes encode nuclear receptors. The osr2 is expressed in the anterior part, while foxP4 in its distal part. Also, we reported the expression pattern of pdgfra for the first time in the larval zebrafish gut. Our data provide fundamental knowledge for studying vertebrate gut regionalization and motility by live imaging using zebrafish.
Collapse
Affiliation(s)
| | - Ayaka Shirai
- School of Science, University of Hyogo, Ako-gun, Japan
| | | | - Shuji Shigenobu
- Trans-Scale Biology Center, National Institute for Basic Biology, Okazaki, Japan
| | - Naoto Ueno
- Trans-Scale Biology Center, National Institute for Basic Biology, Okazaki, Japan
- Unit of Quantitative and Imaging Biology, International Research Collaboration Center, National Institute of Natural Sciences, Okazaki, Japan
| | - Kohei Hatta
- Graduate School of Science, University of Hyogo, Ako-gun, Japan
| |
Collapse
|
14
|
Lin Y, Wu TY, Chen X, Wan S, Chao B, Xin J, Yang JY, Wong WH, Wang YXR. scTIE: data integration and inference of gene regulation using single-cell temporal multimodal data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.18.541381. [PMID: 37292801 PMCID: PMC10245711 DOI: 10.1101/2023.05.18.541381] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Single-cell technologies offer unprecedented opportunities to dissect gene regulatory mechanisms in context-specific ways. Although there are computational methods for extracting gene regulatory relationships from scRNA-seq and scATAC-seq data, the data integration problem, essential for accurate cell type identification, has been mostly treated as a standalone challenge. Here we present scTIE, a unified method that integrates temporal multimodal data and infers regulatory relationships predictive of cellular state changes. scTIE uses an autoencoder to embed cells from all time points into a common space using iterative optimal transport, followed by extracting interpretable information to predict cell trajectories. Using a variety of synthetic and real temporal multimodal datasets, we demonstrate scTIE achieves effective data integration while preserving more biological signals than existing methods, particularly in the presence of batch effects and noise. Furthermore, on the exemplar multiome dataset we generated from differentiating mouse embryonic stem cells over time, we demonstrate scTIE captures regulatory elements highly predictive of cell transition probabilities, providing new potentials to understand the regulatory landscape driving developmental processes.
Collapse
Affiliation(s)
- Yingxin Lin
- School of Mathematics and Statistics, The University of Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, NSW, Australia
- Laboratory of Data Discovery for Health Limited (D24H), Science Park, Hong Kong SAR, China
| | - Tung-Yu Wu
- Department of Statistics, Stanford University, CA, USA
| | - Xi Chen
- Department of Statistics, Stanford University, CA, USA
| | - Sheng Wan
- Institute of Electronics, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Brian Chao
- Department of Electrical Engineering, Stanford University, CA, USA
| | - Jingxue Xin
- Department of Statistics, Stanford University, CA, USA
| | - Jean Y.H. Yang
- School of Mathematics and Statistics, The University of Sydney, NSW, Australia
- Charles Perkins Centre, The University of Sydney, NSW, Australia
- Laboratory of Data Discovery for Health Limited (D24H), Science Park, Hong Kong SAR, China
| | - Wing H. Wong
- Department of Statistics, Stanford University, CA, USA
- Department of Biomedical Data Science, Stanford University, CA, USA
- Bio-X Program, Stanford University, CA, USA
| | - Y. X. Rachel Wang
- School of Mathematics and Statistics, The University of Sydney, NSW, Australia
| |
Collapse
|
15
|
Yang YS, Liu MH, Yan ZW, Chen GQ, Huang Y. FAM122A Is Required for Mesendodermal and Cardiac Differentiation of Embryonic Stem Cells. Stem Cells 2023; 41:354-367. [PMID: 36715298 PMCID: PMC10498146 DOI: 10.1093/stmcls/sxad008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 12/16/2022] [Indexed: 01/31/2023]
Abstract
Mesendodermal specification and cardiac differentiation are key issues for developmental biology and heart regeneration medicine. Previously, we demonstrated that FAM122A, a highly conserved housekeeping gene, is an endogenous inhibitor of protein phosphatase 2A (PP2A) and participates in multifaceted physiological and pathological processes. However, the in vivo function of FAM122A is largely unknown. In this study, we observed that Fam122 deletion resulted in embryonic lethality with severe defects of cardiovascular developments and significantly attenuated cardiac functions in conditional cardiac-specific knockout mice. More importantly, Fam122a deficiency impaired mesendodermal specification and cardiac differentiation from mouse embryonic stem cells but showed no influence on pluripotent identity. Mechanical investigation revealed that the impaired differentiation potential was caused by the dysregulation of histone modification and Wnt and Hippo signaling pathways through modulation of PP2A activity. These findings suggest that FAM122A is a novel and critical regulator in mesendodermal specification and cardiac differentiation. This research not only significantly extends our understanding of the regulatory network of mesendodermal/cardiac differentiation but also proposes the potential significance of FAM122A in cardiac regeneration.
Collapse
Affiliation(s)
- Yun-Sheng Yang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education and Chinese Academy of Medical Sciences Research Unit (2019RU043, Stress and Tumor), Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, People’s Republic of China
| | - Man-Hua Liu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education and Chinese Academy of Medical Sciences Research Unit (2019RU043, Stress and Tumor), Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, People’s Republic of China
| | - Zhao-Wen Yan
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education and Chinese Academy of Medical Sciences Research Unit (2019RU043, Stress and Tumor), Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, People’s Republic of China
| | - Guo-Qiang Chen
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education and Chinese Academy of Medical Sciences Research Unit (2019RU043, Stress and Tumor), Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, People’s Republic of China
| | - Ying Huang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education and Chinese Academy of Medical Sciences Research Unit (2019RU043, Stress and Tumor), Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, People’s Republic of China
| |
Collapse
|
16
|
Downs KM. The mouse allantois: new insights at the embryonic-extraembryonic interface. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210251. [PMID: 36252214 PMCID: PMC9574631 DOI: 10.1098/rstb.2021.0251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 01/20/2022] [Indexed: 12/23/2022] Open
Abstract
During the early development of Placentalia, a distinctive projection emerges at the posterior embryonic-extraembryonic interface of the conceptus; its fingerlike shape presages maturation into the placental umbilical cord, whose major role is to shuttle fetal blood to and from the chorion for exchange with the mother during pregnancy. Until recently, the biology of the cord's vital vascular anlage, called the body stalk/allantois in humans and simply the allantois in rodents, has been largely unknown. Here, new insights into the development of the mouse allantois are featured, from its origin and mechanism of arterial patterning through its union with the chorion. Key to generating the allantois and its critical functions are the primitive streak and visceral endoderm, which together are sufficient to create the entire fetal-placental connection. Their newly discovered roles at the embryonic-extraembryonic interface challenge conventional wisdom, including the physical limits of the primitive streak, its function as sole purveyor of mesoderm in the mouse, potency of visceral endoderm, and the putative role of the allantois in the germ line. With this working model of allantois development, understanding a plethora of hitherto poorly understood orphan diseases in humans is now within reach. This article is part of the theme issue 'Extraembryonic tissues: exploring concepts, definitions and functions across the animal kingdom'.
Collapse
Affiliation(s)
- Karen M. Downs
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison School of Medicine and Public Health, 1111 Highland Avenue, Madison, WI 53705, USA
| |
Collapse
|
17
|
Qin Y, Huang X, Cai Z, Cai B, He J, Yao Y, Zhou C, Kuang J, Yang Y, Chen H, Chen Y, Ou S, Chen L, Wu F, Guo N, Yuan Y, Zhang X, Pang W, Feng Z, Yu S, Liu J, Cao S, Pei D. Regeneration of the human segmentation clock in somitoids in vitro. EMBO J 2022; 41:e110928. [PMID: 36245268 PMCID: PMC9713707 DOI: 10.15252/embj.2022110928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 09/02/2022] [Accepted: 09/16/2022] [Indexed: 01/15/2023] Open
Abstract
Each vertebrate species appears to have a unique timing mechanism for forming somites along the vertebral column, and the process in human remains poorly understood at the molecular level due to technical and ethical limitations. Here, we report the reconstitution of human segmentation clock by direct reprogramming. We first reprogrammed human urine epithelial cells to a presomitic mesoderm (PSM) state capable of long-term self-renewal and formation of somitoids with an anterior-to-posterior axis. By inserting the RNA reporter Pepper into HES7 and MESP2 loci of these iPSM cells, we show that both transcripts oscillate in the resulting somitoids at ~5 h/cycle. GFP-tagged endogenous HES7 protein moves along the anterior-to-posterior axis during somitoid formation. The geo-sequencing analysis further confirmed anterior-to-posterior polarity and revealed the localized expression of WNT, BMP, FGF, and RA signaling molecules and HOXA-D family members. Our study demonstrates the direct reconstitution of human segmentation clock from somatic cells, which may allow future dissection of the mechanism and components of such a clock and aid regenerative medicine.
Collapse
Affiliation(s)
- Yue Qin
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhouChina
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhouChina
- University of the Chinese Academy of SciencesBeijingChina
| | - Xingnan Huang
- Laboratory of Cell Fate Control, School of Life SciencesWestlake UniversityHangzhouChina
| | - Zepo Cai
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhouChina
- Joint School of Life Science, Guangzhou Institutes of Biomedicine and Health, Chinese Academic and SciencesGuangzhou Medical UniversityGuangzhouChina
| | - Baomei Cai
- Center for Cell Lineage and AtlasBioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory)GuangzhouChina
| | - Jiangping He
- Center for Cell Lineage and AtlasBioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory)GuangzhouChina
| | - Yuxiang Yao
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhouChina
| | - Chunhua Zhou
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhouChina
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhouChina
- University of the Chinese Academy of SciencesBeijingChina
| | - Junqi Kuang
- Laboratory of Cell Fate Control, School of Life SciencesWestlake UniversityHangzhouChina
| | - Yihang Yang
- Laboratory of Cell Fate Control, School of Life SciencesWestlake UniversityHangzhouChina
| | - Huan Chen
- Center for Cell Lineage and AtlasBioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory)GuangzhouChina
| | - Yating Chen
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhouChina
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhouChina
- Joint School of Life Science, Guangzhou Institutes of Biomedicine and Health, Chinese Academic and SciencesGuangzhou Medical UniversityGuangzhouChina
| | - Sihua Ou
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhouChina
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhouChina
- Joint School of Life Science, Guangzhou Institutes of Biomedicine and Health, Chinese Academic and SciencesGuangzhou Medical UniversityGuangzhouChina
| | - Lijun Chen
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhouChina
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhouChina
- Joint School of Life Science, Guangzhou Institutes of Biomedicine and Health, Chinese Academic and SciencesGuangzhou Medical UniversityGuangzhouChina
| | - Fang Wu
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhouChina
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhouChina
- University of the Chinese Academy of SciencesBeijingChina
| | - Ning Guo
- Laboratory of Cell Fate Control, School of Life SciencesWestlake UniversityHangzhouChina
| | - Yapei Yuan
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhouChina
| | - Xiangyu Zhang
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhouChina
| | - Wei Pang
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhouChina
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhouChina
| | - Ziyu Feng
- Center for Cell Lineage and AtlasBioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory)GuangzhouChina
| | - Shengyong Yu
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhouChina
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhouChina
- University of the Chinese Academy of SciencesBeijingChina
| | - Jing Liu
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhouChina
- Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and HealthChinese Academy of SciencesGuangzhouChina
- University of the Chinese Academy of SciencesBeijingChina
- Center for Cell Lineage and AtlasBioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory)GuangzhouChina
| | - Shangtao Cao
- Center for Cell Lineage and AtlasBioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory)GuangzhouChina
- Guangzhou LaboratoryGuangzhouChina
| | - Duanqing Pei
- Laboratory of Cell Fate Control, School of Life SciencesWestlake UniversityHangzhouChina
| |
Collapse
|
18
|
Hahner F, Moll F, Warwick T, Hebchen DM, Buchmann GK, Epah J, Abplanalp W, Schader T, Günther S, Gilsbach R, Brandes RP, Schröder K. Nox4 promotes endothelial differentiation through chromatin remodeling. Redox Biol 2022; 55:102381. [PMID: 35810713 PMCID: PMC9287364 DOI: 10.1016/j.redox.2022.102381] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 06/20/2022] [Indexed: 01/09/2023] Open
Abstract
RATIONALE Nox4 is a constitutively active NADPH oxidase that constantly produces low levels of H2O2. Thereby, Nox4 contributes to cell homeostasis and long-term processes, such as differentiation. The high expression of Nox4 seen in endothelial cells contrasts with the low abundance of Nox4 in stem cells, which are accordingly characterized by low levels of H2O2. We hypothesize that Nox4 is a major contributor to endothelial differentiation, is induced during the process of differentiation, and facilitates homeostasis of the resulting endothelial cells. OBJECTIVE To determine the role of No×4 in differentiation of murine inducible pluripotent stem cells (miPSC) into endothelial cells (ECs). METHODS AND RESULTS miPSC, generated from mouse embryonic wildtype (WT) and Nox4-/- fibroblasts, were differentiated into endothelial cells (miPSC-EC) by stimulation with BMP4 and VEGF. During this process, Nox4 expression increased and knockout of Nox4 prolonged the abundance of pluripotency markers, while expression of endothelial markers was delayed in differentiating Nox4-depleted iPSCs. Eventually, angiogenic capacity of iPSC-ECs is reduced in Nox4 deficient cells, indicating that an absence of Nox4 diminishes stability of the reached phenotype. As an underlying mechanism, we identified JmjD3 as a redox target of Nox4. iPSC-ECs lacking Nox4 display a lower nuclear abundance of the histone demethylase JmjD3, resulting in an increased triple methylation of histone 3 (H3K27me3), which serves as a repressive mark for several genes involved in differentiation. CONCLUSIONS Nox4 promotes differentiation of miPSCs into ECs by oxidation of JmjD3 and subsequent demethylation of H3K27me3, which forced endothelial differentiation and stability.
Collapse
Affiliation(s)
- F Hahner
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - F Moll
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - T Warwick
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - D M Hebchen
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - G K Buchmann
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - J Epah
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - W Abplanalp
- Institute for Cardiovascular Regeneration, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - T Schader
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - S Günther
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - R Gilsbach
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - R P Brandes
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - K Schröder
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany.
| |
Collapse
|
19
|
Talbot CD, Walsh MD, Cutty SJ, Elsayed R, Vlachaki E, Bruce AEE, Wardle FC, Nelson AC. Eomes function is conserved between zebrafish and mouse and controls left-right organiser progenitor gene expression via interlocking feedforward loops. Front Cell Dev Biol 2022; 10:982477. [PMID: 36133924 PMCID: PMC9483813 DOI: 10.3389/fcell.2022.982477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
The T-box family transcription factor Eomesodermin (Eomes) is present in all vertebrates, with many key roles in the developing mammalian embryo and immune system. Homozygous Eomes mutant mouse embryos exhibit early lethality due to defects in both the embryonic mesendoderm and the extraembryonic trophoblast cell lineage. In contrast, zebrafish lacking the predominant Eomes homologue A (Eomesa) do not suffer complete lethality and can be maintained. This suggests fundamental differences in either the molecular function of Eomes orthologues or the molecular configuration of processes in which they participate. To explore these hypotheses we initially analysed the expression of distinct Eomes isoforms in various mouse cell types. Next we compared the functional capabilities of these murine isoforms to zebrafish Eomesa. These experiments provided no evidence for functional divergence. Next we examined the functions of zebrafish Eomesa and other T-box family members expressed in early development, as well as its paralogue Eomesb. Though Eomes is a member of the Tbr1 subfamily we found evidence for functional redundancy with the Tbx6 subfamily member Tbx16, known to be absent from eutherians. However, Tbx16 does not appear to synergise with Eomesa cofactors Mixl1 and Gata5. Finally, we analysed the ability of Eomesa and other T-box factors to induce zebrafish left-right organiser progenitors (known as dorsal forerunner cells) known to be positively regulated by vgll4l, a gene we had previously shown to be repressed by Eomesa. Here we demonstrate that Eomesa indirectly upregulates vgll4l expression via interlocking feedforward loops, suggesting a role in establishment of left-right asymmetry. Conversely, other T-box factors could not similarly induce left-right organiser progenitors. Overall these findings demonstrate conservation of Eomes molecular function and participation in similar processes, but differential requirements across evolution due to additional co-expressed T-box factors in teleosts, albeit with markedly different molecular capabilities. Our analyses also provide insights into the role of Eomesa in left-right organiser formation in zebrafish.
Collapse
Affiliation(s)
- Conor D. Talbot
- School of Life Sciences, Gibbet Hill Campus, University of Warwick, Coventry, United Kingdom
| | - Mark D. Walsh
- School of Life Sciences, Gibbet Hill Campus, University of Warwick, Coventry, United Kingdom
| | - Stephen J. Cutty
- Randall Centre for Cell and Molecular Biophysics, New Hunt’s House, Guy’s Campus, King’s College London, London, United Kingdom
| | - Randa Elsayed
- Warwick Medical School, Gibbet Hill Campus, University of Warwick, Coventry, United Kingdom
| | - Eirini Vlachaki
- School of Life Sciences, Gibbet Hill Campus, University of Warwick, Coventry, United Kingdom
| | - Ashley E. E. Bruce
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Fiona C. Wardle
- Randall Centre for Cell and Molecular Biophysics, New Hunt’s House, Guy’s Campus, King’s College London, London, United Kingdom
| | - Andrew C. Nelson
- School of Life Sciences, Gibbet Hill Campus, University of Warwick, Coventry, United Kingdom
| |
Collapse
|
20
|
Luo H, Zhang Y, Deng Y, Li L, Sheng Z, Yu Y, Lin Y, Chen X, Feng P. Nxhl Controls Angiogenesis by Targeting VE-PTP Through Interaction With Nucleolin. Front Cell Dev Biol 2021; 9:728821. [PMID: 34733844 PMCID: PMC8558974 DOI: 10.3389/fcell.2021.728821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/08/2021] [Indexed: 11/13/2022] Open
Abstract
Precise regulation of angiogenesis is required for organ development, wound repair, and tumor progression. Here, we identified a novel gene, nxhl (New XingHuo light), that is conserved in vertebrates and that plays a crucial role in vascular integrity and angiogenesis. Bioinformatic analysis uncovered its essential roles in development based on co-expression with several key developmental genes. Knockdown of nxhl in zebrafish causes global and pericardial edema, loss of blood circulation, and vascular defects characterized by both reduced vascularization in intersegmental vessels and decreased sprouting in the caudal vein plexus. The nxhl gene also affects human endothelial cell behavior in vitro. We found that nxhl functions in part by targeting VE-PTP through interaction with NCL (nucleolin). Loss of ptprb (a VE-PTP ortholo) in zebrafish resulted in defects similar to nxhl knockdown. Moreover, nxhl deficiency attenuates tumor invasion and proteins (including VE-PTP and NCL) associated with angiogenesis and EMT. These findings illustrate that nxhl can regulate angiogenesis via a novel nxhl-NCL-VE-PTP axis, providing a new therapeutic target for modulating vascular formation and function, especially for cancer treatment.
Collapse
Affiliation(s)
- Honglin Luo
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China.,Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Yongde Zhang
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Yanfei Deng
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Lequn Li
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Zhaoan Sheng
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yanling Yu
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Yong Lin
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Xiaohan Chen
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Pengfei Feng
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| |
Collapse
|
21
|
Świerczek-Lasek B, Dudka D, Bauer D, Czajkowski T, Ilach K, Streminska W, Kominek A, Piwocka K, Ciemerych MA, Archacka K. Comparison of Differentiation Pattern and WNT/SHH Signaling in Pluripotent Stem Cells Cultured under Different Conditions. Cells 2021; 10:cells10102743. [PMID: 34685722 PMCID: PMC8534321 DOI: 10.3390/cells10102743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 10/01/2021] [Accepted: 10/08/2021] [Indexed: 11/16/2022] Open
Abstract
Pluripotent stem cells (PSCs) are characterized by the ability to self-renew as well as undergo multidirectional differentiation. Culture conditions have a pivotal influence on differentiation pattern. In the current study, we compared the fate of mouse PSCs using two culture media: (1) chemically defined, free of animal reagents, and (2) standard one relying on the serum supplementation. Moreover, we assessed the influence of selected regulators (WNTs, SHH) on PSC differentiation. We showed that the differentiation pattern of PSCs cultured in both systems differed significantly: cells cultured in chemically defined medium preferentially underwent ectodermal conversion while their endo- and mesodermal differentiation was limited, contrary to cells cultured in serum-supplemented medium. More efficient ectodermal differentiation of PSCs cultured in chemically defined medium correlated with higher activity of SHH pathway while endodermal and mesodermal conversion of cells cultured in serum-supplemented medium with higher activity of WNT/JNK pathway. However, inhibition of either canonical or noncanonical WNT pathway resulted in the limitation of endo- and mesodermal conversion of PSCs. In addition, blocking WNT secretion led to the inhibition of PSC mesodermal differentiation, confirming the pivotal role of WNT signaling in this process. In contrast, SHH turned out to be an inducer of PSC ectodermal, not mesodermal differentiation.
Collapse
Affiliation(s)
- Barbara Świerczek-Lasek
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland; (B.Ś.-L.); (D.D.); (D.B.); (T.C.); (K.I.); (W.S.); (M.A.C.)
| | - Damian Dudka
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland; (B.Ś.-L.); (D.D.); (D.B.); (T.C.); (K.I.); (W.S.); (M.A.C.)
| | - Damian Bauer
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland; (B.Ś.-L.); (D.D.); (D.B.); (T.C.); (K.I.); (W.S.); (M.A.C.)
| | - Tomasz Czajkowski
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland; (B.Ś.-L.); (D.D.); (D.B.); (T.C.); (K.I.); (W.S.); (M.A.C.)
| | - Katarzyna Ilach
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland; (B.Ś.-L.); (D.D.); (D.B.); (T.C.); (K.I.); (W.S.); (M.A.C.)
| | - Władysława Streminska
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland; (B.Ś.-L.); (D.D.); (D.B.); (T.C.); (K.I.); (W.S.); (M.A.C.)
| | - Agata Kominek
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland; (A.K.); (K.P.)
| | - Katarzyna Piwocka
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland; (A.K.); (K.P.)
| | - Maria A. Ciemerych
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland; (B.Ś.-L.); (D.D.); (D.B.); (T.C.); (K.I.); (W.S.); (M.A.C.)
| | - Karolina Archacka
- Department of Cytology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland; (B.Ś.-L.); (D.D.); (D.B.); (T.C.); (K.I.); (W.S.); (M.A.C.)
- Correspondence: ; Tel.: +48-22-55-42-203
| |
Collapse
|
22
|
Lim J, Sakai E, Sakurai F, Mizuguchi H. miR-27b antagonizes BMP signaling in early differentiation of human induced pluripotent stem cells. Sci Rep 2021; 11:19820. [PMID: 34615950 PMCID: PMC8494899 DOI: 10.1038/s41598-021-99403-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/15/2021] [Indexed: 11/21/2022] Open
Abstract
Human induced pluripotent stem (hiPS) cells are feasible materials for studying the biological mechanisms underlying human embryogenesis. In early embryogenesis, definitive endoderm and mesoderm are differentiated from their common precursor, mesendoderm. Bone morphogenetic protein (BMP) signaling is responsible for regulating mesendoderm and mesoderm formation. Micro RNAs (miRNAs), short non-coding RNAs, broadly regulate biological processes via post-transcriptional repression. The expression of miR-27b, which is enriched in somatic cells, has been reported to increase through definitive endoderm and hepatic differentiation, but little is known about how miR-27b acts during early differentiation. Here, we used miR-27b-inducible hiPS cells to investigate the roles of miR-27b in the undifferentiated and early-differentiated stages. In undifferentiated hiPS cells, miR-27b suppressed the expression of pluripotency markers [alkaline phosphatase (AP) and nanog homeobox (NANOG)] and cell proliferation. Once differentiation began, miR-27b expression repressed phosphorylated SMAD1/5, the mediators of the BMP signaling, throughout definitive endoderm differentiation. Consistent with the above findings, miR-27b overexpression downregulated BMP-induced mesendodermal marker genes [Brachyury, mix paired-like homeobox 1 (MIXL1) and eomesodermin (EOMES)], suggesting that miR-27b had an inhibitory effect on early differentiation. Collectively, our findings revealed a novel antagonistic role of miR-27b in the BMP signaling pathway in the early differentiation of hiPS cells.
Collapse
Affiliation(s)
- Jaeeun Lim
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Eiko Sakai
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Fuminori Sakurai
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan. .,Laboratory of Hepatocyte Regulation, National Institute of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito, Asagi, Ibaraki, Osaka, 567-0085, Japan. .,The Center for Advanced Medical Engineering and Informatics, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan. .,Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka, Japan.
| |
Collapse
|
23
|
Ivanovitch K, Soro-Barrio P, Chakravarty P, Jones RA, Bell DM, Mousavy Gharavy SN, Stamataki D, Delile J, Smith JC, Briscoe J. Ventricular, atrial, and outflow tract heart progenitors arise from spatially and molecularly distinct regions of the primitive streak. PLoS Biol 2021; 19:e3001200. [PMID: 33999917 PMCID: PMC8158918 DOI: 10.1371/journal.pbio.3001200] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 05/27/2021] [Accepted: 03/23/2021] [Indexed: 12/22/2022] Open
Abstract
The heart develops from 2 sources of mesoderm progenitors, the first and second heart field (FHF and SHF). Using a single-cell transcriptomic assay combined with genetic lineage tracing and live imaging, we find the FHF and SHF are subdivided into distinct pools of progenitors in gastrulating mouse embryos at earlier stages than previously thought. Each subpopulation has a distinct origin in the primitive streak. The first progenitors to leave the primitive streak contribute to the left ventricle, shortly after right ventricle progenitor emigrate, followed by the outflow tract and atrial progenitors. Moreover, a subset of atrial progenitors are gradually incorporated in posterior locations of the FHF. Although cells allocated to the outflow tract and atrium leave the primitive streak at a similar stage, they arise from different regions. Outflow tract cells originate from distal locations in the primitive streak while atrial progenitors are positioned more proximally. Moreover, single-cell RNA sequencing demonstrates that the primitive streak cells contributing to the ventricles have a distinct molecular signature from those forming the outflow tract and atrium. We conclude that cardiac progenitors are prepatterned within the primitive streak and this prefigures their allocation to distinct anatomical structures of the heart. Together, our data provide a new molecular and spatial map of mammalian cardiac progenitors that will support future studies of heart development, function, and disease.
Collapse
|
24
|
Bardot ES, Hadjantonakis AK. Mouse gastrulation: Coordination of tissue patterning, specification and diversification of cell fate. Mech Dev 2020; 163:103617. [PMID: 32473204 PMCID: PMC7534585 DOI: 10.1016/j.mod.2020.103617] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/18/2020] [Accepted: 05/22/2020] [Indexed: 12/22/2022]
Abstract
During mouse embryonic development a mass of pluripotent epiblast tissue is transformed during gastrulation to generate the three definitive germ layers: endoderm, mesoderm, and ectoderm. During gastrulation, a spatiotemporally controlled sequence of events results in the generation of organ progenitors and positions them in a stereotypical fashion throughout the embryo. Key to the correct specification and differentiation of these cell fates is the establishment of an axial coordinate system along with the integration of multiple signals by individual epiblast cells to produce distinct outcomes. These signaling domains evolve as the anterior-posterior axis is established and the embryo grows in size. Gastrulation is initiated at the posteriorly positioned primitive streak, from which nascent mesoderm and endoderm progenitors ingress and begin to diversify. Advances in technology have facilitated the elaboration of landmark findings that originally described the epiblast fate map and signaling pathways required to execute those fates. Here we will discuss the current state of the field and reflect on how our understanding has shifted in recent years.
Collapse
Affiliation(s)
- Evan S Bardot
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| | - Anna-Katerina Hadjantonakis
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
25
|
Yeo GHT, Lin L, Qi CY, Cha M, Gifford DK, Sherwood RI. A Multiplexed Barcodelet Single-Cell RNA-Seq Approach Elucidates Combinatorial Signaling Pathways that Drive ESC Differentiation. Cell Stem Cell 2020; 26:938-950.e6. [PMID: 32459995 PMCID: PMC7398619 DOI: 10.1016/j.stem.2020.04.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 12/10/2019] [Accepted: 04/27/2020] [Indexed: 01/08/2023]
Abstract
Empirical optimization of stem cell differentiation protocols is time consuming, is laborintensive, and typically does not comprehensively interrogate all relevant signaling pathways. Here we describe barcodelet single-cell RNA sequencing (barRNA-seq), which enables systematic exploration of cellular perturbations by tagging individual cells with RNA "barcodelets" to identify them on the basis of the treatments they receive. We apply barRNA-seq to simultaneously manipulate up to seven developmental pathways and study effects on embryonic stem cell (ESC) germ layer specification and mesodermal specification, uncovering combinatorial effects of signaling pathway activation on gene expression. We further develop a data-driven framework for identifying combinatorial signaling perturbations that drive cells toward specific fates, including several annotated in an existing scRNA-seq gastrulation atlas, and use this approach to guide ESC differentiation into a notochord-like population. We expect that barRNA-seq will have broad utility for investigating and understanding how cooperative signaling pathways drive cell fate acquisition.
Collapse
Affiliation(s)
- Grace Hui Ting Yeo
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Computational and Systems Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Lin Lin
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Hubrecht Institute, 3584 CT Utrecht, the Netherlands
| | - Celine Yueyue Qi
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Minsun Cha
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - David K Gifford
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Richard I Sherwood
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Hubrecht Institute, 3584 CT Utrecht, the Netherlands.
| |
Collapse
|
26
|
Multifactorial Modeling Reveals a Dominant Role of Wnt Signaling in Lineage Commitment of Human Pluripotent Stem Cells. Bioengineering (Basel) 2019; 6:bioengineering6030071. [PMID: 31443254 PMCID: PMC6783940 DOI: 10.3390/bioengineering6030071] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/08/2019] [Accepted: 08/13/2019] [Indexed: 12/17/2022] Open
Abstract
The human primed pluripotent state is maintained by a complex balance of several signaling pathways governing pluripotency maintenance and commitment. Here, we explore a multiparameter approach using a full factorial design and a simple well-defined culture system to assess individual and synergistic contributions of Wnt, FGF and TGFβ signaling to pluripotency and lineage specification of human induced pluripotent stem cells (hiPSC). Hierarchical clustering and quadratic models highlighted a dominant effect of Wnt signaling over FGF and TGFβ signaling, drawing hiPSCs towards mesendoderm lineages. In addition, a synergistic effect between Wnt signaling and FGF was observed to have a negative contribution to pluripotency maintenance and a positive contribution to ectoderm and mesoderm commitment. Furthermore, FGF and TGFβ signaling only contributed significantly for negative ectoderm scores, suggesting that the effect of both factors for pluripotency maintenance resides in a balance of inhibitory signals instead of proactive stimulation of hiPSC pluripotency. Overall, our dry-signaling multiparameter modeling approach can contribute to elucidate individual and synergistic inputs, providing an additional degree of comprehension of the complex regulatory mechanisms of human pluripotency and commitment.
Collapse
|
27
|
Nowotschin S, Hadjantonakis AK, Campbell K. The endoderm: a divergent cell lineage with many commonalities. Development 2019; 146:146/11/dev150920. [PMID: 31160415 PMCID: PMC6589075 DOI: 10.1242/dev.150920] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The endoderm is a progenitor tissue that, in humans, gives rise to the majority of internal organs. Over the past few decades, genetic studies have identified many of the upstream signals specifying endoderm identity in different model systems, revealing them to be divergent from invertebrates to vertebrates. However, more recent studies of the cell behaviours driving endodermal morphogenesis have revealed a surprising number of shared features, including cells undergoing epithelial-to-mesenchymal transitions (EMTs), collective cell migration, and mesenchymal-to-epithelial transitions (METs). In this Review, we highlight how cross-organismal studies of endoderm morphogenesis provide a useful perspective that can move our understanding of this fascinating tissue forward.
Collapse
Affiliation(s)
- Sonja Nowotschin
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anna-Katerina Hadjantonakis
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Kyra Campbell
- Bateson Centre, Firth Court, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK .,Department of Biomedical Science, Firth Court, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| |
Collapse
|
28
|
Genome-scale screens identify JNK-JUN signaling as a barrier for pluripotency exit and endoderm differentiation. Nat Genet 2019; 51:999-1010. [PMID: 31110351 PMCID: PMC6545159 DOI: 10.1038/s41588-019-0408-9] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 04/01/2019] [Indexed: 12/21/2022]
Abstract
Human embryonic and induced pluripotent stem cells (hESCs/hiPSCs) hold great promise for cell-based therapies and drug discovery. However, homogeneous differentiation remains a major challenge, highlighting the need for understanding developmental mechanisms. We performed genome-scale CRISPR screens to uncover regulators of definitive endoderm (DE) differentiation, which unexpectedly uncovered five JNK/JUN family genes as key barriers of DE differentiation. The JNK/JUN pathway does not act through directly inhibiting the DE enhancers. Instead JUN co-occupies ESC enhancers with OCT4, NANOG and SMAD2/3, and specifically inhibits the exit from the pluripotent state by impeding the decommissioning of ESC enhancers and inhibiting the reconfiguration of SMAD2/3 chromatin binding from ESC to DE enhancers. Therefore, the JNK/JUN pathway safeguards pluripotency from precocious DE differentiation. Direct pharmacological inhibition of JNK significantly improves the efficiencies of generating DE and DE-derived pancreatic and lung progenitor cells, highlighting the potential of harnessing the knowledge from developmental studies for regenerative medicine.
Collapse
|
29
|
The role of autophagy in morphogenesis and stem cell maintenance. Histochem Cell Biol 2018; 150:721-732. [PMID: 30382373 DOI: 10.1007/s00418-018-1751-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2018] [Indexed: 12/21/2022]
Abstract
During embryonic development, cells need to undergo a number of morphological changes that are decisive for the shaping of the embryo's body, initiating organogenesis and differentiation into functional tissues. These remodeling processes are accompanied by profound changes in the cell membrane, the cytoskeleton, organelles, and extracellular matrix composition. While considerably detailed insight into the role of autophagy in stem cells biology has been gained in the recent years, information regarding the participation of autophagy in morphogenetic processes is only sparse. This review, therefore, focuses on the role of autophagy in cell morphogenesis through its regulatory activity in TGFβ signaling, expression of adhesion molecules and cell cycle modification. It also discusses the role of autophagy in stem cell maintenance which is very fundamental for cell renewal and replacement during development, pathogenesis of certain diseases and development of therapies. We are thus addressing here perspectives for further potentially rewarding research topics.
Collapse
|
30
|
Blake LE, Thomas SM, Blischak JD, Hsiao CJ, Chavarria C, Myrthil M, Gilad Y, Pavlovic BJ. A comparative study of endoderm differentiation in humans and chimpanzees. Genome Biol 2018; 19:162. [PMID: 30322406 PMCID: PMC6191992 DOI: 10.1186/s13059-018-1490-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 07/20/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND There is substantial interest in the evolutionary forces that shaped the regulatory framework in early human development. Progress in this area has been slow because it is difficult to obtain relevant biological samples. Induced pluripotent stem cells (iPSCs) may provide the ability to establish in vitro models of early human and non-human primate developmental stages. RESULTS Using matched iPSC panels from humans and chimpanzees, we comparatively characterize gene regulatory changes through a four-day time course differentiation of iPSCs into primary streak, endoderm progenitors, and definitive endoderm. As might be expected, we find that differentiation stage is the major driver of variation in gene expression levels, followed by species. We identify thousands of differentially expressed genes between humans and chimpanzees in each differentiation stage. Yet, when we consider gene-specific dynamic regulatory trajectories throughout the time course, we find that at least 75% of genes, including nearly all known endoderm developmental markers, have similar trajectories in the two species. Interestingly, we observe a marked reduction of both intra- and inter-species variation in gene expression levels in primitive streak samples compared to the iPSCs, with a recovery of regulatory variation in endoderm progenitors. CONCLUSIONS The reduction of variation in gene expression levels at a specific developmental stage, paired with overall high degree of conservation of temporal gene regulation, is consistent with the dynamics of a conserved developmental process.
Collapse
Affiliation(s)
- Lauren E. Blake
- Department of Human Genetics, University of Chicago, Chicago, IL USA
| | | | - John D. Blischak
- Department of Human Genetics, University of Chicago, Chicago, IL USA
| | | | - Claudia Chavarria
- Department of Human Genetics, University of Chicago, Chicago, IL USA
| | - Marsha Myrthil
- Department of Human Genetics, University of Chicago, Chicago, IL USA
| | - Yoav Gilad
- Department of Human Genetics, University of Chicago, Chicago, IL USA
- Department of Medicine, University of Chicago, Chicago, IL USA
- Cummings Life Sciences Center, 920 E. 58th Street, CLSC 317, Chicago, IL 60637 USA
| | - Bryan J. Pavlovic
- Department of Human Genetics, University of Chicago, Chicago, IL USA
| |
Collapse
|
31
|
Yokobayashi S, Okita K, Nakagawa M, Nakamura T, Yabuta Y, Yamamoto T, Saitou M. Clonal variation of human induced pluripotent stem cells for induction into the germ cell fate. Biol Reprod 2018; 96:1154-1166. [PMID: 28453617 DOI: 10.1093/biolre/iox038] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 04/26/2017] [Indexed: 02/06/2023] Open
Abstract
The mechanisms for human germ cell development have remained largely unknown, due to the difficulty in obtaining suitable experimental materials. The establishment of an in vitro system to reconstitute human germ cell development will thus provide a critical opportunity to understand its mechanisms at a molecular level. It has previously been shown that human induced pluripotent stem cells (hiPSCs) are first induced into incipient mesoderm-like cells (iMeLCs), which are in turn induced into primordial germ-cell like cells (PGCLCs) with gene expression properties similar to early migratory PGCs. Here, we report that the efficiency of PGCLC induction varies among hiPSC clones, and, interestingly, the clonal variations in PGCLC induction efficiency are reflected in the gene expression states of the iMeLCs. Remarkably, the expression levels of EOMES, MIXL1, or T in the iMeLCs are positively correlated with the efficiency of subsequent PGCLC generation, while the expressions of CDH1, SOX3, or FGF2 are negatively correlated. These results indicate that the expression changes of these genes occurring during iMeLC induction are key markers indicative of successful induction of PGCLCs, and furthermore, that hiPSC clones have different properties that influence their responsivity to the iMeLC induction. Our study thus provides important insights into the mechanism of hPGC specification as well as the development of a better strategy for inducing human germ cell fate from PSCs in vitro.
Collapse
Affiliation(s)
- Shihori Yokobayashi
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, Japan.,Japan Science and Technology Agency, Exploratory Research for Advanced Technology, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, Japan.,Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Keisuke Okita
- Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Masato Nakagawa
- Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, Japan
| | - Tomonori Nakamura
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, Japan.,Japan Science and Technology Agency, Exploratory Research for Advanced Technology, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, Japan
| | - Yukihiro Yabuta
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, Japan.,Japan Science and Technology Agency, Exploratory Research for Advanced Technology, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, Japan
| | - Takuya Yamamoto
- Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, Japan.,Institute for Integrated Cell-Material Sciences, Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku, Kyoto, Japan.,Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology, Japan Agency for Medical Research and Development, Otemachi 1-7-1, Chiyoda-ku, Tokyo, Japan
| | - Mitinori Saitou
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, Japan.,Japan Science and Technology Agency, Exploratory Research for Advanced Technology, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, Japan.,Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, Japan.,Institute for Integrated Cell-Material Sciences, Kyoto University, Yoshida-Ushinomiya-cho, Sakyo-ku, Kyoto, Japan
| |
Collapse
|
32
|
Wang Y, Zhou T, Wan J, Yang Y, Chen X, Wang J, Zhou C, Liu M, Ling X, Zhang J. Comparative transcriptome analysis reveals a regulatory network of microRNA-29b during mouse early embryonic development. Oncotarget 2018; 7:53772-53782. [PMID: 27449102 PMCID: PMC5288220 DOI: 10.18632/oncotarget.10741] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 07/09/2016] [Indexed: 12/27/2022] Open
Abstract
MicroRNAs are endogenous ~22 nt RNAs that regulate gene expression by translational inhibition and mRNA destabilization. MicroRNA-29b (miR-29b) is essential for progression of mouse embryos past preimplantation development; however, details of the underlying regulatory network remain to be elucidated. Here, we used RNA sequencing to identify changes in the transcriptome of mouse embryos in response to miR-29b inhibition. Morula-stage embryos that had been subject to miR-29b inhibition throughout preimplantation development exhibited significant expression changes in 870 genes compared with controls. Among 405 genes that were downregulated, 30 genes encoded factors with known essential function during early embryonic development, including the pluripotent stem cell factor Nanog. We identified 19 genes encoding putative miR-29b target transcripts. These included Zbtb40, Hbp1, Ccdc117, Ypel2, Klf4, and Tmed9, which are upregulated at the 4-cell state of mouse development concomitant with miR-29b downregulation. Luciferase reporter analysis confirmed that Zbtb40, Hbp1, Ccdc117, Ypel2, and Klf4 transcripts are direct targets of miR-29b. These results suggest that miR-29b decreases the mRNA levels of several target genes during early mouse development, including the gene encoding the reprogramming factor Klf4. We hypothesize that inhibition of miR-29b causes overexpression of its target genes, triggering downstream signaling networks to decrease the expression of genes that are essential for embryonic development. In conclusion, miR-29b controls an extensive regulatory network in early mouse embryos, which comprises reprogramming factors and molecular regulators of post-transcriptional modification processes.
Collapse
Affiliation(s)
- Ying Wang
- Department of Reproduction, Nanjing Maternity and Child Health Care Hospital, Affiliated to Nanjing Medical University, Nanjing 210004, P.R. China
| | - Tao Zhou
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing 210029, P.R. China
| | - Jinyuan Wan
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing 210029, P.R. China
| | - Ye Yang
- Department of Reproduction, Nanjing Maternity and Child Health Care Hospital, Affiliated to Nanjing Medical University, Nanjing 210004, P.R. China
| | - Xiaojiao Chen
- Department of Reproduction, Nanjing Maternity and Child Health Care Hospital, Affiliated to Nanjing Medical University, Nanjing 210004, P.R. China
| | - Jiayi Wang
- Department of Reproduction, Nanjing Maternity and Child Health Care Hospital, Affiliated to Nanjing Medical University, Nanjing 210004, P.R. China
| | - Cheng Zhou
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing 210029, P.R. China
| | - Mingxi Liu
- State Key Laboratory of Reproductive Medicine, Department of Histology and Embryology, Nanjing Medical University, Nanjing 210029, P.R. China
| | - Xiufeng Ling
- Department of Reproduction, Nanjing Maternity and Child Health Care Hospital, Affiliated to Nanjing Medical University, Nanjing 210004, P.R. China
| | - Junqiang Zhang
- Department of Reproduction, Nanjing Maternity and Child Health Care Hospital, Affiliated to Nanjing Medical University, Nanjing 210004, P.R. China
| |
Collapse
|
33
|
Shirouzu Y, Yanai G, Yang KC, Sumi S. Effects of Activin in Embryoid Bodies Expressing Fibroblast Growth Factor 5. Cell Reprogram 2017; 18:171-86. [PMID: 27253628 DOI: 10.1089/cell.2015.0074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Nodal/activin signaling is indispensable for embryonic development. We examined what activin does to the embryoid bodies (EBs) produced from mouse embryonic stem cells (mESCs) expressing an epiblast marker. The EBs were produced by culturing mESCs by the hanging drop method for 24 hours. The resulting EBs were transferred onto gelatin-coated dishes and allowed to further differentiate. The 24-hour EBs showed a stronger expression of fibroblast growth factor (FGF)5 and Brachyury (specific to the epiblast) in comparison with mESCs. Treating the transferred EBs with activin A maintained transcript levels of FGF5 and Oct4, while inhibiting definitive endoderm differentiation. The activin A treatment reversed the endoderm differentiation induced by retinoic acid (RA), while the inhibition of nodal/activin signaling promoted RA-induced endoderm differentiation. Inhibition of nodal/activin signaling in EBs, including epiblast-like cells, promotes differentiation into the endoderm, facilitating the transition from the pluripotent state to specification of the endoderm.
Collapse
Affiliation(s)
- Yasumasa Shirouzu
- Department of Organ Reconstruction, Institute for Frontier Medical Sciences, Kyoto University , Kyoto, Japan
| | - Goichi Yanai
- Department of Organ Reconstruction, Institute for Frontier Medical Sciences, Kyoto University , Kyoto, Japan
| | - Kai-Chiang Yang
- Department of Organ Reconstruction, Institute for Frontier Medical Sciences, Kyoto University , Kyoto, Japan
| | - Shoichiro Sumi
- Department of Organ Reconstruction, Institute for Frontier Medical Sciences, Kyoto University , Kyoto, Japan
| |
Collapse
|
34
|
Yu S, Liao J, Tang M, Wang Y, Wei X, Mao L, Zeng C, Wang G. A functional single nucleotide polymorphism in the tyrosinase gene promoter affects skin color and transcription activity in the black-boned chicken. Poult Sci 2017; 96:4061-4067. [DOI: 10.3382/ps/pex217] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 08/22/2017] [Indexed: 12/14/2022] Open
|
35
|
Evolutionarily Distinctive Transcriptional and Signaling Programs Drive Human Germ Cell Lineage Specification from Pluripotent Stem Cells. Cell Stem Cell 2017; 21:517-532.e5. [DOI: 10.1016/j.stem.2017.09.005] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 07/24/2017] [Accepted: 09/07/2017] [Indexed: 12/21/2022]
|
36
|
Gene expression analysis of bovine embryonic disc, trophoblast and parietal hypoblast at the start of gastrulation. ZYGOTE 2017; 25:265-278. [PMID: 28534463 DOI: 10.1017/s0967199417000090] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
In cattle early gastrulation-stage embryos (Stage 5), four tissues can be discerned: (i) the top layer of the embryonic disc consisting of embryonic ectoderm (EmE); (ii) the bottom layer of the disc consisting of mesoderm, endoderm and visceral hypoblast (MEH); (iii) the trophoblast (TB); and (iv) the parietal hypoblast. We performed microsurgery followed by RNA-seq to analyse the transcriptome of these four tissues as well as a developmentally earlier pre-gastrulation embryonic disc. The cattle EmE transcriptome was similar at Stages 4 and 5, characterised by the OCT4/SOX2/NANOG pluripotency network. Expression of genes associated with primordial germ cells suggest their presence in the EmE tissue at these stages. Anterior visceral hypoblast genes were transcribed in the Stage 4 disc, but no longer by Stage 5. The Stage 5 MEH layer was equally similar to mouse embryonic and extraembryonic visceral endoderm. Our data suggest that the first mesoderm to invaginate in cattle embryos is fated to become extraembryonic. TGFβ, FGF, VEGF, PDGFA, IGF2, IHH and WNT signals and receptors were expressed, however the representative members of the FGF families differed from that seen in equivalent tissues of mouse embryos. The TB transcriptome was unique and differed significantly from that of mice. FGF signalling in the TB may be autocrine with both FGFR2 and FGF2 expressed. Our data revealed a range of potential inter-tissue interactions, highlighted significant differences in early development between mice and cattle and yielded insight into the developmental events occurring at the start of gastrulation.
Collapse
|
37
|
Jang S, Choubey S, Furchtgott L, Zou LN, Doyle A, Menon V, Loew EB, Krostag AR, Martinez RA, Madisen L, Levi BP, Ramanathan S. Dynamics of embryonic stem cell differentiation inferred from single-cell transcriptomics show a series of transitions through discrete cell states. eLife 2017; 6:20487. [PMID: 28296635 PMCID: PMC5352225 DOI: 10.7554/elife.20487] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 01/31/2017] [Indexed: 02/06/2023] Open
Abstract
The complexity of gene regulatory networks that lead multipotent cells to acquire different cell fates makes a quantitative understanding of differentiation challenging. Using a statistical framework to analyze single-cell transcriptomics data, we infer the gene expression dynamics of early mouse embryonic stem (mES) cell differentiation, uncovering discrete transitions across nine cell states. We validate the predicted transitions across discrete states using flow cytometry. Moreover, using live-cell microscopy, we show that individual cells undergo abrupt transitions from a naïve to primed pluripotent state. Using the inferred discrete cell states to build a probabilistic model for the underlying gene regulatory network, we further predict and experimentally verify that these states have unique response to perturbations, thus defining them functionally. Our study provides a framework to infer the dynamics of differentiation from single cell transcriptomics data and to build predictive models of the gene regulatory networks that drive the sequence of cell fate decisions during development. DOI:http://dx.doi.org/10.7554/eLife.20487.001
Collapse
Affiliation(s)
- Sumin Jang
- FAS Center for Systems Biology, Harvard University, Cambridge, United States.,Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States
| | - Sandeep Choubey
- FAS Center for Systems Biology, Harvard University, Cambridge, United States.,Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States
| | - Leon Furchtgott
- FAS Center for Systems Biology, Harvard University, Cambridge, United States.,Biophysics Program, Harvard University, Cambridge, United States
| | - Ling-Nan Zou
- FAS Center for Systems Biology, Harvard University, Cambridge, United States
| | - Adele Doyle
- FAS Center for Systems Biology, Harvard University, Cambridge, United States.,Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States
| | - Vilas Menon
- Allen Institute for Brain Science, Seattle, United States
| | - Ethan B Loew
- FAS Center for Systems Biology, Harvard University, Cambridge, United States.,Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States
| | | | | | - Linda Madisen
- Allen Institute for Brain Science, Seattle, United States
| | - Boaz P Levi
- Allen Institute for Brain Science, Seattle, United States
| | - Sharad Ramanathan
- FAS Center for Systems Biology, Harvard University, Cambridge, United States.,Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States.,Allen Institute for Brain Science, Seattle, United States.,School of Engineering and Applied Sciences, Harvard University, Cambridge, United States.,Harvard Stem Cell Institute, Harvard University, Cambridge, United States
| |
Collapse
|
38
|
Stem Cell Technology in Cardiac Regeneration: A Pluripotent Stem Cell Promise. EBioMedicine 2017; 16:30-40. [PMID: 28169191 PMCID: PMC5474503 DOI: 10.1016/j.ebiom.2017.01.029] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 01/13/2017] [Accepted: 01/20/2017] [Indexed: 12/21/2022] Open
Abstract
Despite advances in cardiovascular biology and medical therapy, heart disorders are the leading cause of death worldwide. Cell-based regenerative therapies become a promising treatment for patients affected by heart failure, but also underline the need for reproducible results in preclinical and clinical studies for safety and efficacy. Enthusiasm has been tempered by poor engraftment, survival and differentiation of the injected adult stem cells. The crucial challenge is identification and selection of the most suitable stem cell type for cardiac regenerative medicine. Human pluripotent stem cells (PSCs) have emerged as attractive cell source to obtain cardiomyocytes (CMs), with potential applications, including drug discovery and toxicity screening, disease modelling and innovative cell therapies. Lessons from embryology offered important insights into the development of stem cell-derived CMs. However, the generation of a CM population, uniform in cardiac subtype, adult maturation and functional properties, is highly recommended. Moreover, hurdles regarding tumorigenesis, graft cell death, immune rejection and arrhythmogenesis need to be overcome in clinical practice. Here we highlight the recent progression in PSC technologies for the regeneration of injured heart. We review novel strategies that might overcome current obstacles in heart regenerative medicine, aiming at improving cell survival and functional integration after cell transplantation. Human pluripotent stem cells emerge as attractive tool for cardiac regeneration approaches. Plasticity of human pluripotent stem cells towards cardiac-related cell types guarantees repopulation of injured heart. Combination of stem cell and gene editing therapies has potential to become next generation treatment for cardiac diseases.
Data for this Review were identified by searches of MEDLINE and PubMed, and references from relevant articles using the search terms “cardiomyogenesis”, “adult stem cells”, “pluripotent stem cells” and “cardiac regeneration”. Only articles published in English between 1976 and 2017 were included. The majority of the articles reported were published after 2000.
Collapse
|
39
|
Takasato M, Little MH. A strategy for generating kidney organoids: Recapitulating the development in human pluripotent stem cells. Dev Biol 2016; 420:210-220. [PMID: 27565022 PMCID: PMC6186756 DOI: 10.1016/j.ydbio.2016.08.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 06/19/2016] [Accepted: 08/21/2016] [Indexed: 02/06/2023]
Abstract
Directed differentiation of human pluripotent stem cells (hPSCs) can provide us any required tissue/cell types by recapitulating the development in vitro. The kidney is one of the most challenging organs to generate from hPSCs as the kidney progenitors are composed of at least 4 different cell types, including nephron, collecting duct, endothelial and interstitium progenitors, that are developmentally distinguished populations. Although the actual developmental process of the kidney during human embryogenesis has not been clarified yet, studies using model animals accumulated knowledge about the origins of kidney progenitors. The implications of these findings for the directed differentiation of hPSCs into the kidney include the mechanism of the intermediate mesoderm specification and its patterning along with anteroposterior axis. Using this knowledge, we previously reported successful generation of hPSCs-derived kidney organoids that contained all renal components and modelled human kidney development in vitro. In this review, we explain the developmental basis of the strategy behind this differentiation protocol and compare strategies of studies that also recently reported the induction of kidney cells from hPSCs. We also discuss the characterization of such kidney organoids and limitations and future applications of this technology.
Collapse
Affiliation(s)
- Minoru Takasato
- Murdoch Childrens Research Institute, Parkville, Victoria 3052, Australia; RIKEN Center for Developmental Biology, Kobe 650-0047, Japan.
| | - Melissa H Little
- Murdoch Childrens Research Institute, Parkville, Victoria 3052, Australia; Department of Pediatrics, University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
40
|
Yang P, Chen X, Kaushal S, Reece EA, Yang P. High glucose suppresses embryonic stem cell differentiation into cardiomyocytes : High glucose inhibits ES cell cardiogenesis. Stem Cell Res Ther 2016; 7:187. [PMID: 27938398 PMCID: PMC5148851 DOI: 10.1186/s13287-016-0446-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 11/11/2016] [Accepted: 11/22/2016] [Indexed: 12/18/2022] Open
Abstract
Background Babies born to mothers with pregestational diabetes have a high risk for congenital heart defects (CHD). Embryonic stem cells (ESCs) are excellent in vitro models for studying the effect of high glucose on cardiac lineage specification because ESCs can be differentiated into cardiomyocytes. ESC maintenance and differentiation are currently performed under high glucose conditions, whose adverse effects have never been clarified. Method We investigated the effect of high glucose on cardiomyocyte differentiation from a well-characterized ESC line, E14, derived from mouse blastocysts. E14 cells maintained under high glucose (25 mM) failed to generate any beating cardiomyocytes using the hanging-drop embryonic body method. We created a glucose-responsive E14 cell line (GR-E14) through a graduated low glucose adaptation. The expression of stem cell markers was similar in the parent E14 cells and the GR-E14 cells. Results Glucose transporter 2 gene was increased in GR-E14 cells. When GR-E14 cells were differentiated into cardiomyocytes under low (5 mM) or high (25 mM) glucose conditions, high glucose significantly delayed the appearance and reduced the number of TNNT2 (Troponin T Type 2)-positive contracting cardiomyocytes. High glucose suppressed the expression of precardiac mesoderm markers, cardiac transcription factors, mature cardiomyocyte markers, and potassium channel proteins. High glucose impaired the functionality of ESC-derived cardiomyocytes by suppressing the frequencies of Ca2+ wave and contraction. Conclusions Our findings suggest that high glucose inhibits ESC cardiogenesis by suppressing key developmental genes essential for the cardiac program. Electronic supplementary material The online version of this article (doi:10.1186/s13287-016-0446-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Penghua Yang
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, BRB11-039, 655W. Baltimore Street, Baltimore, MD, 21201, USA
| | - Xi Chen
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, BRB11-039, 655W. Baltimore Street, Baltimore, MD, 21201, USA
| | - Sunjay Kaushal
- Division of Cardiac Surgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - E Albert Reece
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, BRB11-039, 655W. Baltimore Street, Baltimore, MD, 21201, USA.,Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Peixin Yang
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, BRB11-039, 655W. Baltimore Street, Baltimore, MD, 21201, USA. .,Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
41
|
Abstract
The treatment of renal failure has seen little change in the past 70 years. Patients with end-stage renal disease (ESRD) are treated with renal replacement therapy, including dialysis or organ transplantation. The growing imbalance between the availability of donor organs and prevalence of ESRD is pushing an increasing number of patients to undergo dialysis. Although the prospect of new treatment options for patients through regenerative medicine has long been suggested, advances in the generation of human kidney cell types through the directed differentiation of human pluripotent stem cells over the past 2 years have brought this prospect closer to delivery. These advances are the result of careful research into mammalian embryogenesis. By understanding the decision points made within the embryo to pattern the kidney, it is now possible to recreate self-organizing kidney tissues in vitro. In this Review, we describe the key decision points in kidney development and how these decisions have been mimicked experimentally. Recreation of human nephrons from human pluripotent stem cells opens the door to patient-derived disease models and personalized drug and toxicity screening. In the long term, we hope that these efforts will also result in the generation of bioengineered organs for the treatment of kidney disease.
Collapse
|
42
|
Jin JZ, Zhu Y, Warner D, Ding J. Analysis of extraembryonic mesodermal structure formation in the absence of morphological primitive streak. Dev Growth Differ 2016; 58:522-9. [PMID: 27273137 DOI: 10.1111/dgd.12294] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 04/15/2016] [Accepted: 04/17/2016] [Indexed: 11/30/2022]
Abstract
During mouse gastrulation, the primitive streak is formed on the posterior side of the embryo. Cells migrate out of the primitive streak to form the future mesoderm and endoderm. Fate mapping studies revealed a group of cell migrate through the proximal end of the primitive streak and give rise to the extraembryonic mesoderm tissues such as the yolk sac blood islands and allantois. However, it is not clear whether the formation of a morphological primitive streak is required for the development of these extraembryonic mesodermal tissues. Loss of the Cripto gene in mice dramatically reduces, but does not completely abolish, Nodal activity leading to the absence of a morphological primitive streak. However, embryonic erythrocytes are still formed and assembled into the blood islands. In addition, Cripto mutant embryos form allantoic buds. However, Drap1 mutant embryos have excessive Nodal activity in the epiblast cells before gastrulation and form an expanded primitive streak, but no yolk sac blood islands or allantoic bud formation. Lefty2 embryos also have elevated levels of Nodal activity in the primitive streak during gastrulation, and undergo normal blood island and allantois formation. We therefore speculate that low level of Nodal activity disrupts the formation of morphological primitive streak on the posterior side, but still allows the formation of primitive streak cells on the proximal side, which give rise to the extraembryonic mesodermal tissues formation. Excessive Nodal activity in the epiblast at pre-gastrulation stage, but not in the primitive streak cells during gastrulation, disrupts extraembryonic mesoderm development.
Collapse
Affiliation(s)
- Jiu-Zhen Jin
- Department of Molecular, Cellular & Craniofacial Biology and Birth Defects Center, University of Louisville, Louisville, Kentucky, USA
| | - Yuanqi Zhu
- Department of Molecular, Cellular & Craniofacial Biology and Birth Defects Center, University of Louisville, Louisville, Kentucky, USA
| | - Dennis Warner
- Department of Molecular, Cellular & Craniofacial Biology and Birth Defects Center, University of Louisville, Louisville, Kentucky, USA
| | - Jixiang Ding
- Department of Molecular, Cellular & Craniofacial Biology and Birth Defects Center, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
43
|
Bell CC, Amaral PP, Kalsbeek A, Magor GW, Gillinder KR, Tangermann P, di Lisio L, Cheetham SW, Gruhl F, Frith J, Tallack MR, Ru KL, Crawford J, Mattick JS, Dinger ME, Perkins AC. The Evx1/Evx1as gene locus regulates anterior-posterior patterning during gastrulation. Sci Rep 2016; 6:26657. [PMID: 27226347 PMCID: PMC4880930 DOI: 10.1038/srep26657] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/29/2016] [Indexed: 01/09/2023] Open
Abstract
Thousands of sense-antisense mRNA-lncRNA gene pairs occur in the mammalian genome. While there is usually little doubt about the function of the coding transcript, the function of the lncRNA partner is mostly untested. Here we examine the function of the homeotic Evx1-Evx1as gene locus. Expression is tightly co-regulated in posterior mesoderm of mouse embryos and in embryoid bodies. Expression of both genes is enhanced by BMP4 and WNT3A, and reduced by Activin. We generated a suite of deletions in the locus by CRISPR-Cas9 editing. We show EVX1 is a critical downstream effector of BMP4 and WNT3A with respect to patterning of posterior mesoderm. The lncRNA, Evx1as arises from alternative promoters and is difficult to fully abrogate by gene editing or siRNA approaches. Nevertheless, we were able to generate a large 2.6 kb deletion encompassing the shared promoter with Evx1 and multiple additional exons of Evx1as. This led to an identical dorsal-ventral patterning defect to that generated by micro-deletion in the DNA-binding domain of EVX1. Thus, Evx1as has no function independent of EVX1, and is therefore unlikely to act in trans. We predict many antisense lncRNAs have no specific trans function, possibly only regulating the linked coding genes in cis.
Collapse
Affiliation(s)
- Charles C Bell
- Mater Research, Translational Research Institute, University of Queensland, Brisbane, Queensland, 4101, Australia
| | - Paulo P Amaral
- The Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Anton Kalsbeek
- The Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia.,Garvan Institute of Medical Research, Sydney, Australia
| | - Graham W Magor
- Mater Research, Translational Research Institute, University of Queensland, Brisbane, Queensland, 4101, Australia
| | - Kevin R Gillinder
- Mater Research, Translational Research Institute, University of Queensland, Brisbane, Queensland, 4101, Australia
| | - Pierre Tangermann
- The Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Lorena di Lisio
- Mater Research, Translational Research Institute, University of Queensland, Brisbane, Queensland, 4101, Australia
| | - Seth W Cheetham
- Mater Research, Translational Research Institute, University of Queensland, Brisbane, Queensland, 4101, Australia.,Diamantina Institute; Translational Research Institute, University of Queensland, Brisbane, Queensland, 4102, Australia
| | - Franziska Gruhl
- Mater Research, Translational Research Institute, University of Queensland, Brisbane, Queensland, 4101, Australia.,Diamantina Institute; Translational Research Institute, University of Queensland, Brisbane, Queensland, 4102, Australia
| | - Jessica Frith
- The Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia.,The Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, 4102, Australia
| | - Michael R Tallack
- Mater Research, Translational Research Institute, University of Queensland, Brisbane, Queensland, 4101, Australia.,The Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Ke-Lin Ru
- The Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia.,The Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, 4102, Australia
| | - Joanna Crawford
- The Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - John S Mattick
- Garvan Institute of Medical Research, Sydney, Australia.,St Vincents Clinical School, Faculty of Medicine, UNSW Australia, Sydney, Australia
| | - Marcel E Dinger
- Garvan Institute of Medical Research, Sydney, Australia.,Diamantina Institute; Translational Research Institute, University of Queensland, Brisbane, Queensland, 4102, Australia.,St Vincents Clinical School, Faculty of Medicine, UNSW Australia, Sydney, Australia
| | - Andrew C Perkins
- Mater Research, Translational Research Institute, University of Queensland, Brisbane, Queensland, 4101, Australia.,The Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia.,The Princess Alexandra Hospital, Brisbane, Queensland, 4102, Australia
| |
Collapse
|
44
|
Johnson J, Lee W, Frazier AE, Vaghjiani V, Laskowski A, Rodriguez AL, Cagnone GL, McKenzie M, White SJ, Nisbet DR, Thorburn DR, St. John JC. Deletion of the Complex I Subunit NDUFS4 Adversely Modulates Cellular Differentiation. Stem Cells Dev 2016; 25:239-50. [DOI: 10.1089/scd.2015.0211] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Jacqueline Johnson
- Centre for Genetic Diseases, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Australia
| | - William Lee
- Centre for Genetic Diseases, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Australia
| | - Ann E. Frazier
- Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, Australia
- Department of Pediatrics, University of Melbourne, Melbourne, Australia
| | - Vijesh Vaghjiani
- Centre for Genetic Diseases, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Australia
| | - Adrienne Laskowski
- Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, Australia
| | | | - Gael L. Cagnone
- Centre for Genetic Diseases, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Australia
| | - Matthew McKenzie
- Centre for Genetic Diseases, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Australia
| | - Stefan J. White
- Centre for Genetic Diseases, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Australia
| | - David R. Nisbet
- Research School of Engineering, Australian National University, Canberra, Australia
| | - David R. Thorburn
- Murdoch Childrens Research Institute, Royal Children's Hospital, Melbourne, Australia
- Department of Pediatrics, University of Melbourne, Melbourne, Australia
| | - Justin C. St. John
- Centre for Genetic Diseases, Hudson Institute of Medical Research, Clayton, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Australia
| |
Collapse
|
45
|
Świerczek B, Ciemerych MA, Archacka K. From pluripotency to myogenesis: a multistep process in the dish. J Muscle Res Cell Motil 2015; 36:363-75. [PMID: 26715014 PMCID: PMC4762919 DOI: 10.1007/s10974-015-9436-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 11/30/2015] [Indexed: 12/11/2022]
Abstract
Pluripotent stem cells (PSCs), such as embryonic stem cells or induced pluripotent stem cells are a promising source of cells for regenerative medicine as they can differentiate into all cell types building a mammalian body. However, protocols leading to efficient and safe in vitro generation of desired cell types must be perfected before PSCs can be used in cell therapies or tissue engineering. In vivo, i.e. in developing mouse embryo or teratoma, PSCs can differentiate into skeletal muscle, but in vitro their spontaneous differentiation into myogenic cells is inefficient. Numerous attempts have been undertaken to enhance this process. Many of them involved mimicking the interactions occurring during embryonic myogenesis. The key regulators of embryonic myogenesis, such as Wnts proteins, fibroblast growth factor 2, and retinoic acid, have been tested to improve the frequency of in vitro myogenic differentiation of PSCs. This review summarizes the current state of the art, comparing spontaneous and directed myogenic differentiation of PSCs as well as the protocols developed this far to facilitate this process.
Collapse
Affiliation(s)
- Barbara Świerczek
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096, Warsaw, Poland
| | - Maria A Ciemerych
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096, Warsaw, Poland
| | - Karolina Archacka
- Department of Cytology, Faculty of Biology, University of Warsaw, Miecznikowa 1, 02-096, Warsaw, Poland.
| |
Collapse
|
46
|
Novel Mode of Defective Neural Tube Closure in the Non-Obese Diabetic (NOD) Mouse Strain. Sci Rep 2015; 5:16917. [PMID: 26593875 PMCID: PMC4655353 DOI: 10.1038/srep16917] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 10/21/2015] [Indexed: 11/08/2022] Open
Abstract
Failure to close the neural tube results in birth defects, with severity ranging from spina bifida to lethal anencephaly. Few genetic risk factors for neural tube defects are known in humans, highlighting the critical role of environmental risk factors, such as maternal diabetes. Yet, it is not well understood how altered maternal metabolism interferes with embryonic development, and with neurulation in particular. We present evidence from two independent mouse models of diabetic pregnancy that identifies impaired migration of nascent mesodermal cells in the primitive streak as the morphogenetic basis underlying the pathogenesis of neural tube defects. We conclude that perturbed gastrulation not only explains the neurulation defects, but also provides a unifying etiology for the broad spectrum of congenital malformations in diabetic pregnancies.
Collapse
|
47
|
Chen G, Tan R, Tao Q. Sebox regulates mesoderm formation in early amphibian embryos. Dev Dyn 2015; 244:1415-26. [DOI: 10.1002/dvdy.24323] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2015] [Revised: 08/02/2015] [Accepted: 08/04/2015] [Indexed: 01/02/2023] Open
Affiliation(s)
- Geng Chen
- MOE Key Laboratory of Protein Sciences; Tsinghua University School of Life Sciences; Beijing China
| | - Renbo Tan
- MOE Key Laboratory of Protein Sciences; Tsinghua University School of Life Sciences; Beijing China
| | - Qinghua Tao
- MOE Key Laboratory of Protein Sciences; Tsinghua University School of Life Sciences; Beijing China
| |
Collapse
|
48
|
Palpant NJ, Pabon L, Roberts M, Hadland B, Jones D, Jones C, Moon RT, Ruzzo WL, Bernstein I, Zheng Y, Murry CE. Inhibition of β-catenin signaling respecifies anterior-like endothelium into beating human cardiomyocytes. Development 2015; 142:3198-209. [PMID: 26153229 PMCID: PMC4582173 DOI: 10.1242/dev.117010] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 05/26/2015] [Indexed: 01/14/2023]
Abstract
During vertebrate development, mesodermal fate choices are regulated by interactions between morphogens such as activin/nodal, BMPs and Wnt/β-catenin that define anterior-posterior patterning and specify downstream derivatives including cardiomyocyte, endothelial and hematopoietic cells. We used human embryonic stem cells to explore how these pathways control mesodermal fate choices in vitro. Varying doses of activin A and BMP4 to mimic cytokine gradient polarization in the anterior-posterior axis of the embryo led to differential activity of Wnt/β-catenin signaling and specified distinct anterior-like (high activin/low BMP) and posterior-like (low activin/high BMP) mesodermal populations. Cardiogenic mesoderm was generated under conditions specifying anterior-like mesoderm, whereas blood-forming endothelium was generated from posterior-like mesoderm, and vessel-forming CD31+ endothelial cells were generated from all mesoderm origins. Surprisingly, inhibition of β-catenin signaling led to the highly efficient respecification of anterior-like endothelium into beating cardiomyocytes. Cardiac respecification was not observed in posterior-derived endothelial cells. Thus, activin/BMP gradients specify distinct mesodermal subpopulations that generate cell derivatives with unique angiogenic, hemogenic and cardiogenic properties that should be useful for understanding embryogenesis and developing therapeutics. Summary: The manipulation of signals that control embryonic patterning allows human pluripotent stem cells to be differentiated into endothelial subpopulations with distinct haematopoietic, angiogenic and cardiogenic potential.
Collapse
Affiliation(s)
- Nathan J Palpant
- Department of Pathology, University of Washington School of Medicine, Seattle, WA 98109, USA Center for Cardiovascular Biology, University of Washington School of Medicine, Seattle, WA 98109, USA Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Lil Pabon
- Department of Pathology, University of Washington School of Medicine, Seattle, WA 98109, USA Center for Cardiovascular Biology, University of Washington School of Medicine, Seattle, WA 98109, USA Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Meredith Roberts
- Center for Cardiovascular Biology, University of Washington School of Medicine, Seattle, WA 98109, USA Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA Department of Bioengineering, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Brandon Hadland
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98109, USA Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Daniel Jones
- Department of Computer Science and Engineering, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Christina Jones
- Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98109, USA Howard Hughes Medical Institute, Seattle, WA 98109, USA
| | - Randall T Moon
- Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98109, USA Howard Hughes Medical Institute, Seattle, WA 98109, USA
| | - Walter L Ruzzo
- Department of Computer Science and Engineering, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Irwin Bernstein
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98109, USA Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Ying Zheng
- Center for Cardiovascular Biology, University of Washington School of Medicine, Seattle, WA 98109, USA Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA Department of Bioengineering, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Charles E Murry
- Department of Pathology, University of Washington School of Medicine, Seattle, WA 98109, USA Center for Cardiovascular Biology, University of Washington School of Medicine, Seattle, WA 98109, USA Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA Department of Bioengineering, University of Washington School of Medicine, Seattle, WA 98109, USA Department of Medicine/Cardiology, University of Washington School of Medicine, Seattle, WA 98109, USA
| |
Collapse
|
49
|
Kaufman-Francis K, Goh HN, Kojima Y, Studdert JB, Jones V, Power MD, Wilkie E, Teber E, Loebel DAF, Tam PPL. Differential response of epiblast stem cells to Nodal and Activin signalling: a paradigm of early endoderm development in the embryo. Philos Trans R Soc Lond B Biol Sci 2015; 369:rstb.2013.0550. [PMID: 25349457 DOI: 10.1098/rstb.2013.0550] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Mouse epiblast stem cells (EpiSCs) display temporal differences in the upregulation of Mixl1 expression during the initial steps of in vitro differentiation, which can be correlated with their propensity for endoderm differentiation. EpiSCs that upregulated Mixl1 rapidly during differentiation responded robustly to both Activin A and Nodal in generating foregut endoderm and precursors of pancreatic and hepatic tissues. By contrast, EpiSCs that delayed Mixl1 upregulation responded less effectively to Nodal and showed an overall suboptimal outcome of directed differentiation. The enhancement in endoderm potency in Mixl1-early cells may be accounted for by a rapid exit from the progenitor state and the efficient response to the induction of differentiation by Nodal. EpiSCs that readily differentiate into the endoderm cells are marked by a distinctive expression fingerprint of transforming growth factor (TGF)-β signalling pathway genes and genes related to the endoderm lineage. Nodal appears to elicit responses that are associated with transition to a mesenchymal phenotype, whereas Activin A promotes gene expression associated with maintenance of an epithelial phenotype. We postulate that the formation of definitive endoderm (DE) in embryoid bodies follows a similar process to germ layer formation from the epiblast, requiring an initial de-epithelialization event and subsequent re-epithelialization. Our results show that priming EpiSCs with the appropriate form of TGF-β signalling at the formative phase of endoderm differentiation impacts on the further progression into mature DE-derived lineages, and that this is influenced by the initial characteristics of the cell population. Our study also highlights that Activin A, which is commonly used as an in vitro surrogate for Nodal in differentiation protocols, does not elicit the same downstream effects as Nodal, and therefore may not effectively mimic events that take place in the mouse embryo.
Collapse
Affiliation(s)
- Keren Kaufman-Francis
- Embryology Unit, Children's Medical Research Institute, Westmead, New South Wales 2145, Australia
| | - Hwee Ngee Goh
- Embryology Unit, Children's Medical Research Institute, Westmead, New South Wales 2145, Australia
| | - Yoji Kojima
- Embryology Unit, Children's Medical Research Institute, Westmead, New South Wales 2145, Australia Institute of Integrated Cell-Material Science, Kyoto University, Kyoto 606-8501, Japan
| | - Joshua B Studdert
- Embryology Unit, Children's Medical Research Institute, Westmead, New South Wales 2145, Australia
| | - Vanessa Jones
- Embryology Unit, Children's Medical Research Institute, Westmead, New South Wales 2145, Australia
| | - Melinda D Power
- Embryology Unit, Children's Medical Research Institute, Westmead, New South Wales 2145, Australia
| | - Emilie Wilkie
- Embryology Unit, Children's Medical Research Institute, Westmead, New South Wales 2145, Australia Bioinformatics Group, Children's Medical Research Institute, Westmead, New South Wales 2145, Australia
| | - Erdahl Teber
- Bioinformatics Group, Children's Medical Research Institute, Westmead, New South Wales 2145, Australia
| | - David A F Loebel
- Embryology Unit, Children's Medical Research Institute, Westmead, New South Wales 2145, Australia Sydney Medical School, University of Sydney, Sydney, New South Wales 2008, Australia
| | - Patrick P L Tam
- Embryology Unit, Children's Medical Research Institute, Westmead, New South Wales 2145, Australia Sydney Medical School, University of Sydney, Sydney, New South Wales 2008, Australia
| |
Collapse
|
50
|
Viotti M, Foley AC, Hadjantonakis AK. Gutsy moves in mice: cellular and molecular dynamics of endoderm morphogenesis. Philos Trans R Soc Lond B Biol Sci 2015; 369:rstb.2013.0547. [PMID: 25349455 DOI: 10.1098/rstb.2013.0547] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Despite the importance of the gut and its accessory organs, our understanding of early endoderm development is still incomplete. Traditionally, endoderm has been difficult to study because of its small size and relative fragility. However, recent advances in live cell imaging technologies have dramatically expanded our understanding of this tissue, adding a new appreciation for the complex molecular and morphogenetic processes that mediate gut formation. Several spatially and molecularly distinct subpopulations have been shown to exist within the endoderm before the onset of gastrulation. Here, we review findings that have uncovered complex cell movements within the endodermal layer, before and during gastrulation, leading to the conclusion that cells from primitive endoderm contribute descendants directly to gut.
Collapse
Affiliation(s)
- Manuel Viotti
- Genentech Incorporated, South San Francisco, CA 94080, USA
| | - Ann C Foley
- Department of Bioengineering, Clemson University, Charleston, SC 29425, USA
| | | |
Collapse
|