1
|
Thowfeequ S, Hanna CW, Srinivas S. Origin, fate and function of extraembryonic tissues during mammalian development. Nat Rev Mol Cell Biol 2025; 26:255-275. [PMID: 39627419 DOI: 10.1038/s41580-024-00809-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/05/2024] [Indexed: 03/28/2025]
Abstract
Extraembryonic tissues have pivotal roles in morphogenesis and patterning of the early mammalian embryo. Developmental programmes mediated through signalling pathways and gene regulatory networks determine the sequence in which fate determination and lineage commitment of extraembryonic tissues take place, and epigenetic processes allow the memory of cell identity and state to be sustained throughout and beyond embryo development, even extending across generations. In this Review, we discuss the molecular and cellular mechanisms necessary for the different extraembryonic tissues to develop and function, from their initial specification up until the end of gastrulation, when the body plan of the embryo and the anatomical organization of its supporting extraembryonic structures are established. We examine the interaction between extraembryonic and embryonic tissues during early patterning and morphogenesis, and outline how epigenetic memory supports extraembryonic tissue development.
Collapse
Affiliation(s)
- Shifaan Thowfeequ
- Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Courtney W Hanna
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Loke Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Shankar Srinivas
- Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK.
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
2
|
Duncan RK, Liu L, Moyer M, Wylie A, Dano R, Cassinotti L. Retinoic acid signaling guides the efficiency of inner ear organoid-genesis and governs sensory-nonsensory fate specification. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.21.644434. [PMID: 40166165 PMCID: PMC11957153 DOI: 10.1101/2025.03.21.644434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Inner ear organoid development-from germ layer to otocyst formation-relies on timed chemical cues to recapitulate major signals in vivo. In contrast, later stages of differentiation-from otic vesicle (OV) to organoid formation-are self-guided, even though these stages are modulated by several key morphogens in vivo . We sought to elucidate additional morphogens that might improve culture efficiency and influence cell fate decisions. Using a whole-transcriptomic approach, we identified major differences in native and stem cell-derived OVs related to anterior-posterior patterning and retinoic acid (RA) signaling. Increasing the level of RA during OV formation in these cultures modulated organoid efficiency, increased nonsensory markers, decreased sensory markers, and decreased hair cell production. The organoid culture platform mimics the exquisite RA sensitivity found in normal inner ear development and may help identify RA-responsive genes driving organogenesis and cell fate specification.
Collapse
Affiliation(s)
- R. Keith Duncan
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
- Department of Otolaryngology – Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI USA
- Veterans Affairs Ann Arbor Health System, Ann Arbor, MI, USA
- Senior author
| | - Liqian Liu
- Department of Otolaryngology – Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI USA
| | - Mo Moyer
- Department of Otolaryngology – Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI USA
| | - Andrew Wylie
- Department of Otolaryngology – Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI USA
| | - Ranya Dano
- Department of Otolaryngology – Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI USA
| | - Luis Cassinotti
- Department of Otolaryngology – Head and Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI USA
| |
Collapse
|
3
|
Cui L, Lin S, Yang X, Xie X, Wang X, He N, Yang J, Zhang X, Lu X, Yan X, Guo Y, Zhang B, Li R, Miao H, Ji M, Zhang R, Yu L, Xiao Z, Wei Y, Guo J. Spatial transcriptomic characterization of a Carnegie stage 7 human embryo. Nat Cell Biol 2025; 27:360-369. [PMID: 39794460 DOI: 10.1038/s41556-024-01597-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 12/12/2024] [Indexed: 01/13/2025]
Abstract
Gastrulation marks a pivotal stage in mammalian embryonic development, establishing the three germ layers and body axis through lineage diversification and morphogenetic movements. However, studying human gastrulating embryos is challenging due to limited access to early tissues. Here we show the use of spatial transcriptomics to analyse a fully intact Carnegie stage 7 human embryo at single-cell resolution, along with immunofluorescence validations in a second embryo. Employing 82 serial cryosections and Stereo-seq technology, we reconstructed a three-dimensional model of the embryo. Our findings reveal early specification of distinct mesoderm subtypes and the presence of the anterior visceral endoderm. Notably, primordial germ cells were located in the connecting stalk, and haematopoietic stem cell-independent haematopoiesis was observed in the yolk sac. This study advances our understanding of human gastrulation and provides a valuable dataset for future research in early human development.
Collapse
Affiliation(s)
- Lina Cui
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Sirui Lin
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing, China
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xiaolong Yang
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Xinwei Xie
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Xiaoyan Wang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Nannan He
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jingyu Yang
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Xin Zhang
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing, China
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xiaojian Lu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Xiaodi Yan
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Yifei Guo
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Bailing Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Ran Li
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Hefan Miao
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Mei Ji
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Runzhao Zhang
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Leqian Yu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
| | - Zhenyu Xiao
- School of Life Science, Beijing Institute of Technology, Beijing, China.
| | - Yulei Wei
- Frontiers Science Center for Molecular Design Breeding (MOE), China Agricultural University, Beijing, China.
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China.
| | - Jingtao Guo
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
4
|
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
|
5
|
Thowfeequ S, Fiorentino J, Hu D, Solovey M, Ruane S, Whitehead M, Zhou F, Godwin J, Mateo-Otero Y, Vanhaesebroeck B, Scialdone A, Srinivas S. An integrated approach identifies the molecular underpinnings of murine anterior visceral endoderm migration. Dev Cell 2024; 59:2347-2363.e9. [PMID: 38843837 PMCID: PMC11511681 DOI: 10.1016/j.devcel.2024.05.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 11/09/2023] [Accepted: 05/14/2024] [Indexed: 09/12/2024]
Abstract
The anterior visceral endoderm (AVE) differs from the surrounding visceral endoderm (VE) in its migratory behavior and ability to restrict primitive streak formation to the opposite side of the mouse embryo. To characterize the molecular bases for the unique properties of the AVE, we combined single-cell RNA sequencing of the VE prior to and during AVE migration with phosphoproteomics, high-resolution live-imaging, and short-term lineage labeling and intervention. This identified the transient nature of the AVE with attenuation of "anteriorizing" gene expression as cells migrate and the emergence of heterogeneities in transcriptional states relative to the AVE's position. Using cell communication analysis, we identified the requirement of semaphorin signaling for normal AVE migration. Lattice light-sheet microscopy showed that Sema6D mutants have abnormalities in basal projections and migration speed. These findings point to a tight coupling between transcriptional state and position of the AVE and identify molecular controllers of AVE migration.
Collapse
Affiliation(s)
- Shifaan Thowfeequ
- Institute for Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7TY, UK
| | - Jonathan Fiorentino
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich 81377, Germany; Institute of Functional Epigenetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany; Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany; Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, Rome 00161, Italy
| | - Di Hu
- Institute for Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7TY, UK
| | - Maria Solovey
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich 81377, Germany; Institute of Functional Epigenetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany; Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany
| | - Sharon Ruane
- Institute for Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7TY, UK
| | - Maria Whitehead
- UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Felix Zhou
- University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jonathan Godwin
- Institute for Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7TY, UK
| | - Yentel Mateo-Otero
- Institute for Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7TY, UK; Unit of Cell Biology, Department of Biology, University of Girona, Girona 17004, Spain
| | | | - Antonio Scialdone
- Institute of Epigenetics and Stem Cells, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich 81377, Germany; Institute of Functional Epigenetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany; Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg 85764, Germany.
| | - Shankar Srinivas
- Institute for Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7TY, UK.
| |
Collapse
|
6
|
Dupont C. A comprehensive review: synergizing stem cell and embryonic development knowledge in mouse and human integrated stem cell-based embryo models. Front Cell Dev Biol 2024; 12:1386739. [PMID: 38715920 PMCID: PMC11074781 DOI: 10.3389/fcell.2024.1386739] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 04/05/2024] [Indexed: 01/06/2025] Open
Abstract
Mammalian stem cell-based embryo models have emerged as innovative tools for investigating early embryogenesis in both mice and primates. They not only reduce the need for sacrificing mice but also overcome ethical limitations associated with human embryo research. Furthermore, they provide a platform to address scientific questions that are otherwise challenging to explore in vivo. The usefulness of a stem cell-based embryo model depends on its fidelity in replicating development, efficiency and reproducibility; all essential for addressing biological queries in a quantitative manner, enabling statistical analysis. Achieving such fidelity and efficiency requires robust systems that demand extensive optimization efforts. A profound understanding of pre- and post-implantation development, cellular plasticity, lineage specification, and existing models is imperative for making informed decisions in constructing these models. This review aims to highlight essential differences in embryo development and stem cell biology between mice and humans, assess how these variances influence the formation of partially and fully integrated stem cell models, and identify critical challenges in the field.
Collapse
Affiliation(s)
- Cathérine Dupont
- Department of Developmental Biology, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
7
|
Perera M, Brickman JM. In vitro models of human hypoblast and mouse primitive endoderm. Curr Opin Genet Dev 2023; 83:102115. [PMID: 37783145 DOI: 10.1016/j.gde.2023.102115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/28/2023] [Accepted: 08/24/2023] [Indexed: 10/04/2023]
Abstract
The primitive endoderm (PrE, also named hypoblast), a predominantly extraembryonic epithelium that arises from the inner cell mass (ICM) of the mammalian pre-implantation blastocyst, plays a fundamental role in embryonic development, giving rise to the yolk sac, establishing the anterior-posterior axis and contributing to the gut. PrE is specified from the ICM at the same time as the epiblast (Epi) that will form the embryo proper. While in vitro cell lines resembling the pluripotent Epi have been derived from a variety of conditions, only one model system currently exists for the PrE, naïve extraembryonic endoderm (nEnd). As a result, considerably more is known about the gene regulatory networks and signalling requirements of pluripotent stem cells than nEnd. In this review, we describe the ontogeny and differentiation of the PrE or hypoblast in mouse and primate and then discuss in vitro cell culture models for different extraembryonic endodermal cell types.
Collapse
Affiliation(s)
- Marta Perera
- reNEW UCPH - The Novo Nordisk Foundation Center for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark. https://twitter.com/@MartaPrera
| | - Joshua M Brickman
- reNEW UCPH - The Novo Nordisk Foundation Center for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark.
| |
Collapse
|
8
|
Cheng T, Xing YY, Liu C, Li YF, Huang Y, Liu X, Zhang YJ, Zhao GQ, Dong Y, Fu XX, Tian YM, Shu LP, Megason SG, Xu PF. Nodal coordinates the anterior-posterior patterning of germ layers and induces head formation in zebrafish explants. Cell Rep 2023; 42:112351. [PMID: 37018074 DOI: 10.1016/j.celrep.2023.112351] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 01/16/2023] [Accepted: 03/21/2023] [Indexed: 04/06/2023] Open
Abstract
Much progress has been made toward generating analogs of early embryos, such as gastruloids and embryoids, in vitro. However, methods for how to fully mimic the cell movements of gastrulation and coordinate germ-layer patterning to induce head formation are still lacking. Here, we show that a regional Nodal gradient applied to zebrafish animal pole explant can generate a structure that recapitulates the key cell movements of gastrulation. Using single-cell transcriptome and in situ hybridization analysis, we assess the dynamics of the cell fates and patterning of this structure. The mesendoderm differentiates into the anterior endoderm, prechordal plate, notochord, and tailbud-like cells along an anterior-posterior axis, and an anterior-posterior-patterned head-like structure (HLS) progressively forms during late gastrulation. Among 105 immediate Nodal targets, 14 genes contain axis-induction ability, and 5 of them induce a complete or partial head structure when overexpressed in the ventral side of zebrafish embryos.
Collapse
Affiliation(s)
- Tao Cheng
- Women's Hospital, Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yan-Yi Xing
- Women's Hospital, Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorders, Hangzhou, Zhejiang, China
| | - Cong Liu
- Women's Hospital, Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yun-Fei Li
- Women's Hospital, Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ying Huang
- Women's Hospital, Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiang Liu
- Women's Hospital, Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ying-Jie Zhang
- Women's Hospital, Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Guo-Qin Zhao
- Department of Immunology, Guizhou Medical University, Guiyang 550004, China
| | - Yang Dong
- Women's Hospital, Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xin-Xin Fu
- Women's Hospital, Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yi-Meng Tian
- Women's Hospital, Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Li-Ping Shu
- Department of Immunology, Guizhou Medical University, Guiyang 550004, China
| | - Sean G Megason
- Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA.
| | - Peng-Fei Xu
- Women's Hospital, Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
9
|
Dupont C, Schäffers OJ, Tan BF, Merzouk S, Bindels EM, Zwijsen A, Huylebroeck D, Gribnau J. Efficient generation of ETX embryoids that recapitulate the entire window of murine egg cylinder development. SCIENCE ADVANCES 2023; 9:eadd2913. [PMID: 36652512 PMCID: PMC9848479 DOI: 10.1126/sciadv.add2913] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 12/20/2022] [Indexed: 06/17/2023]
Abstract
The murine embryonic-trophoblast-extra-embryonic endoderm (ETX) model is an integrated stem cell-based model to study early postimplantation development. It is based on the self-assembly potential of embryonic, trophoblast, and hypoblast/primitive/visceral endoderm-type stem cell lines (ESC, TSC, and XEN, respectively) to arrange into postimplantation egg cylinder-like embryoids. Here, we provide an optimized method for reliable and efficient generation of ETX embryoids that develop into late gastrulation in static culture conditions. It is based on transgenic Gata6-overproducing ESCs and modified assembly and culture conditions. Using this method, up to 43% of assembled ETX embryoids exhibited a correct spatial distribution of the three stem cell derivatives at day 4 of culture. Of those, 40% progressed into ETX embryoids that both transcriptionally and morphologically faithfully mimicked in vivo postimplantation mouse development between E5.5 and E7.5. The ETX model system offers the opportunity to study the murine postimplantation egg cylinder stages and could serve as a source of various cell lineage precursors.
Collapse
Affiliation(s)
- Cathérine Dupont
- Department of Developmental Biology, Erasmus University Medical Center, Rotterdam, Netherlands
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Olivier J. M. Schäffers
- Department of Developmental Biology, Erasmus University Medical Center, Rotterdam, Netherlands
- Department of Obstetrics and Fetal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Beatrice F. Tan
- Department of Developmental Biology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Sarra Merzouk
- Department of Developmental Biology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Eric M. Bindels
- Department of Hematology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - An Zwijsen
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Danny Huylebroeck
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Joost Gribnau
- Department of Developmental Biology, Erasmus University Medical Center, Rotterdam, Netherlands
- Oncode Institute, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
10
|
Zhu Q, Ge J, Liu Y, Xu JW, Yan S, Zhou F. Decoding anterior-posterior axis emergence among mouse, monkey, and human embryos. Dev Cell 2023; 58:63-79.e4. [PMID: 36626872 DOI: 10.1016/j.devcel.2022.12.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 08/23/2022] [Accepted: 12/07/2022] [Indexed: 01/11/2023]
Abstract
Anterior-posterior axis formation regulated by the distal visceral endoderm (DVE) and anterior visceral endoderm (AVE) is essential for peri-implantation embryogenesis. However, the principles of the origin and specialization of DVE and AVE remain elusive. Here, with single-cell transcriptome analysis and pseudotime prediction, we show that DVE and AVE independently originate from the specialized primary endoderm in mouse blastocysts. Along distinct developmental paths, these two lineages, respectively, undergo four representative states with stage-specific transcriptional patterns around implantation. Further comparative analysis shows that AVE, but not DVE, is detected in human and non-human primate embryos, defining differences in polarity formation across species. Moreover, stem cell-assembled human blastoids lack DVE or AVE precursors, implying that additional induction of stem cells with DVE/AVE potential could promote the current embryo-like models and their post-implantation growth. Our work provides insight into understanding of embryonic polarity formation and early mammalian development.
Collapse
Affiliation(s)
- Qingyuan Zhu
- Haihe Laboratory of Cell Ecosystem, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jitao Ge
- Haihe Laboratory of Cell Ecosystem, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Ying Liu
- Haihe Laboratory of Cell Ecosystem, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jia-Wen Xu
- Haihe Laboratory of Cell Ecosystem, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Shengyi Yan
- Haihe Laboratory of Cell Ecosystem, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Fan Zhou
- Haihe Laboratory of Cell Ecosystem, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
11
|
McMahon R, Sibbritt T, Aryamanesh N, Masamsetti VP, Tam PPL. Loss of Foxd4 Impacts Neurulation and Cranial Neural Crest Specification During Early Head Development. Front Cell Dev Biol 2022; 9:777652. [PMID: 35178396 PMCID: PMC8843869 DOI: 10.3389/fcell.2021.777652] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/30/2021] [Indexed: 11/19/2022] Open
Abstract
The specification of anterior head tissue in the late gastrulation mouse embryo relies on signaling cues from the visceral endoderm and anterior mesendoderm (AME). Genetic loss-of-function studies have pinpointed a critical requirement of LIM homeobox 1 (LHX1) transcription factor in these tissues for the formation of the embryonic head. Transcriptome analysis of embryos with gain-of-function LHX1 activity identified the forkhead box gene, Foxd4, as one downstream target of LHX1 in late-gastrulation E7.75 embryos. Our analysis of single-cell RNA-seq data show Foxd4 is co-expressed with Lhx1 and Foxa2 in the anterior midline tissue of E7.75 mouse embryos, and in the anterior neuroectoderm (ANE) at E8.25 alongside head organizer genes Otx2 and Hesx1. To study the role of Foxd4 during early development we used CRISPR-Cas9 gene editing in mouse embryonic stem cells (mESCs) to generate bi-allelic frameshift mutations in the coding sequence of Foxd4. In an in vitro model of the anterior neural tissues derived from Foxd4-loss of function (LOF) mESCs and extraembryonic endoderm cells, expression of head organizer genes as well as Zic1 and Zic2 was reduced, pointing to a need for FOXD4 in regulating early neuroectoderm development. Mid-gestation mouse chimeras harbouring Foxd4-LOF mESCs displayed craniofacial malformations and neural tube closure defects. Furthermore, our in vitro data showed a loss of FOXD4 impacts the expression of cranial neural crest markers Twist1 and Sox9. Our findings have demonstrated that FOXD4 is essential in the AME and later in the ANE for rostral neural tube closure and neural crest specification during head development.
Collapse
Affiliation(s)
- Riley McMahon
- Embryology Research Unit, Children's Medical Research Institute, Sydney, NSW, Australia.,School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Darlington, NSW, Australia
| | - Tennille Sibbritt
- Embryology Research Unit, Children's Medical Research Institute, Sydney, NSW, Australia
| | - Nadar Aryamanesh
- Embryology Research Unit, Children's Medical Research Institute, Sydney, NSW, Australia
| | - V Pragathi Masamsetti
- Embryology Research Unit, Children's Medical Research Institute, Sydney, NSW, Australia.,School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Darlington, NSW, Australia
| | - Patrick P L Tam
- Embryology Research Unit, Children's Medical Research Institute, Sydney, NSW, Australia.,School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Darlington, NSW, Australia
| |
Collapse
|
12
|
Ueda Y, Kimura-Yoshida C, Mochida K, Tsume M, Kameo Y, Adachi T, Lefebvre O, Hiramatsu R, Matsuo I. Intrauterine Pressures Adjusted by Reichert's Membrane Are Crucial for Early Mouse Morphogenesis. Cell Rep 2021; 31:107637. [PMID: 32433954 DOI: 10.1016/j.celrep.2020.107637] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/10/2020] [Accepted: 04/21/2020] [Indexed: 10/24/2022] Open
Abstract
Mammalian embryogenesis proceeds in utero with the support of nutrients and gases from maternal tissues. However, the contribution of the mechanical environment provided by the uterus to embryogenesis remains unaddressed. Notably, how intrauterine pressures are produced, accurately adjusted, and exerted on embryos are completely unknown. Here, we find that Reichert's membrane, a specialized basement membrane that wraps around the implanted mouse embryo, plays a crucial role as a shock absorber to protect embryos from intrauterine pressures. Notably, intrauterine pressures are produced by uterine smooth muscle contractions, showing the highest and most frequent periodic peaks just after implantation. Mechanistically, such pressures are adjusted within the sealed space between the embryo and uterus created by Reichert's membrane and are involved in egg-cylinder morphogenesis as an important biomechanical environment in utero. Thus, we propose the buffer space sealed by Reichert's membrane cushions and disperses intrauterine pressures exerted on embryos for egg-cylinder morphogenesis.
Collapse
Affiliation(s)
- Yoko Ueda
- Department of Molecular Embryology, Research Institute, Osaka Women's and Children's Hospital, Osaka Prefectural Hospital Organization, 840, Murodo-cho, Izumi, Osaka 594-1101, Japan
| | - Chiharu Kimura-Yoshida
- Department of Molecular Embryology, Research Institute, Osaka Women's and Children's Hospital, Osaka Prefectural Hospital Organization, 840, Murodo-cho, Izumi, Osaka 594-1101, Japan
| | - Kyoko Mochida
- Department of Molecular Embryology, Research Institute, Osaka Women's and Children's Hospital, Osaka Prefectural Hospital Organization, 840, Murodo-cho, Izumi, Osaka 594-1101, Japan
| | - Mami Tsume
- Department of Molecular Embryology, Research Institute, Osaka Women's and Children's Hospital, Osaka Prefectural Hospital Organization, 840, Murodo-cho, Izumi, Osaka 594-1101, Japan
| | - Yoshitaka Kameo
- Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Taiji Adachi
- Institute for Frontier Life and Medical Sciences, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Olivier Lefebvre
- INSERM UMR_S1109, Université de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg 67000, France
| | - Ryuji Hiramatsu
- Department of Molecular Embryology, Research Institute, Osaka Women's and Children's Hospital, Osaka Prefectural Hospital Organization, 840, Murodo-cho, Izumi, Osaka 594-1101, Japan
| | - Isao Matsuo
- Department of Molecular Embryology, Research Institute, Osaka Women's and Children's Hospital, Osaka Prefectural Hospital Organization, 840, Murodo-cho, Izumi, Osaka 594-1101, Japan.
| |
Collapse
|
13
|
Wang X, Xiang Y, Yu Y, Wang R, Zhang Y, Xu Q, Sun H, Zhao ZA, Jiang X, Wang X, Lu X, Qin D, Quan Y, Zhang J, Shyh-Chang N, Wang H, Jing N, Xie W, Li L. Formative pluripotent stem cells show features of epiblast cells poised for gastrulation. Cell Res 2021; 31:526-541. [PMID: 33608671 PMCID: PMC8089102 DOI: 10.1038/s41422-021-00477-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 01/22/2021] [Indexed: 01/29/2023] Open
Abstract
The pluripotency of mammalian early and late epiblast could be recapitulated by naïve embryonic stem cells (ESCs) and primed epiblast stem cells (EpiSCs), respectively. However, these two states of pluripotency may not be sufficient to reflect the full complexity and developmental potency of the epiblast during mammalian early development. Here we report the establishment of self-renewing formative pluripotent stem cells (fPSCs) which manifest features of epiblast cells poised for gastrulation. fPSCs can be established from different mouse ESCs, pre-/early-gastrula epiblasts and induced PSCs. Similar to pre-/early-gastrula epiblasts, fPSCs show the transcriptomic features of formative pluripotency, which are distinct from naïve ESCs and primed EpiSCs. fPSCs show the unique epigenetic states of E6.5 epiblast, including the super-bivalency of a large set of developmental genes. Just like epiblast cells immediately before gastrulation, fPSCs can efficiently differentiate into three germ layers and primordial germ cells (PGCs) in vitro. Thus, fPSCs highlight the feasibility of using PSCs to explore the development of mammalian epiblast.
Collapse
Affiliation(s)
- Xiaoxiao Wang
- grid.410726.60000 0004 1797 8419State Key Laboratory of Stem Cell and Reproductive Biology, Innovation Academy for Stem Cell and Regeneration, Beijing Institute for Stem Cell and Regenerative Medicine, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101 China
| | - Yunlong Xiang
- grid.203458.80000 0000 8653 0555Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016 China
| | - Yang Yu
- grid.410726.60000 0004 1797 8419State Key Laboratory of Stem Cell and Reproductive Biology, Innovation Academy for Stem Cell and Regeneration, Beijing Institute for Stem Cell and Regenerative Medicine, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101 China
| | - Ran Wang
- grid.9227.e0000000119573309State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Yu Zhang
- grid.12527.330000 0001 0662 3178Center for Stem Cell Biology and Regenerative Medicine, MOE Key Laboratory of Bioinformatics, THU-PKU Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084 China
| | - Qianhua Xu
- grid.12527.330000 0001 0662 3178Center for Stem Cell Biology and Regenerative Medicine, MOE Key Laboratory of Bioinformatics, THU-PKU Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084 China
| | - Hao Sun
- grid.410726.60000 0004 1797 8419State Key Laboratory of Stem Cell and Reproductive Biology, Innovation Academy for Stem Cell and Regeneration, Beijing Institute for Stem Cell and Regenerative Medicine, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101 China
| | - Zhen-Ao Zhao
- grid.410726.60000 0004 1797 8419State Key Laboratory of Stem Cell and Reproductive Biology, Innovation Academy for Stem Cell and Regeneration, Beijing Institute for Stem Cell and Regenerative Medicine, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101 China
| | - Xiangxiang Jiang
- grid.410726.60000 0004 1797 8419State Key Laboratory of Stem Cell and Reproductive Biology, Innovation Academy for Stem Cell and Regeneration, Beijing Institute for Stem Cell and Regenerative Medicine, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101 China
| | - Xiaoqing Wang
- grid.410726.60000 0004 1797 8419State Key Laboratory of Stem Cell and Reproductive Biology, Innovation Academy for Stem Cell and Regeneration, Beijing Institute for Stem Cell and Regenerative Medicine, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101 China
| | - Xukun Lu
- grid.410726.60000 0004 1797 8419State Key Laboratory of Stem Cell and Reproductive Biology, Innovation Academy for Stem Cell and Regeneration, Beijing Institute for Stem Cell and Regenerative Medicine, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101 China
| | - Dandan Qin
- grid.410726.60000 0004 1797 8419State Key Laboratory of Stem Cell and Reproductive Biology, Innovation Academy for Stem Cell and Regeneration, Beijing Institute for Stem Cell and Regenerative Medicine, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101 China
| | - Yujun Quan
- grid.410726.60000 0004 1797 8419State Key Laboratory of Stem Cell and Reproductive Biology, Innovation Academy for Stem Cell and Regeneration, Beijing Institute for Stem Cell and Regenerative Medicine, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101 China
| | - Jiaqi Zhang
- grid.410726.60000 0004 1797 8419State Key Laboratory of Stem Cell and Reproductive Biology, Innovation Academy for Stem Cell and Regeneration, Beijing Institute for Stem Cell and Regenerative Medicine, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101 China
| | - Ng Shyh-Chang
- grid.410726.60000 0004 1797 8419State Key Laboratory of Stem Cell and Reproductive Biology, Innovation Academy for Stem Cell and Regeneration, Beijing Institute for Stem Cell and Regenerative Medicine, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101 China
| | - Hongmei Wang
- grid.410726.60000 0004 1797 8419State Key Laboratory of Stem Cell and Reproductive Biology, Innovation Academy for Stem Cell and Regeneration, Beijing Institute for Stem Cell and Regenerative Medicine, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101 China
| | - Naihe Jing
- grid.9227.e0000000119573309State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, 200031 China ,grid.9227.e0000000119573309Guangzhou Regenerative Medicine and Health Guangdong Laboratory (GRMH-GDL), Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong 510530 China
| | - Wei Xie
- grid.12527.330000 0001 0662 3178Center for Stem Cell Biology and Regenerative Medicine, MOE Key Laboratory of Bioinformatics, THU-PKU Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084 China
| | - Lei Li
- grid.410726.60000 0004 1797 8419State Key Laboratory of Stem Cell and Reproductive Biology, Innovation Academy for Stem Cell and Regeneration, Beijing Institute for Stem Cell and Regenerative Medicine, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101 China
| |
Collapse
|
14
|
Gastruloids generated without exogenous Wnt activation develop anterior neural tissues. Stem Cell Reports 2021; 16:1143-1155. [PMID: 33891872 PMCID: PMC8185432 DOI: 10.1016/j.stemcr.2021.03.017] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 03/17/2021] [Accepted: 03/17/2021] [Indexed: 11/21/2022] Open
Abstract
When stimulated with a pulse from an exogenous WNT pathway activator, small aggregates of mouse embryonic stem cells (ESCs) can undergo embryo-like axial morphogenesis and patterning along the three major body axes. However, these structures, called gastruloids, currently lack the anterior embryonic regions, such as those belonging to the brain. Here, we describe an approach to generate gastruloids that have a more complete antero-posterior development. We used hydrogel microwell arrays to promote the robust derivation of mouse ESCs into post-implantation epiblast-like (EPI) aggregates in a reproducible and scalable manner. These EPI aggregates break symmetry and axially elongate without external chemical stimulation. Inhibition of WNT signaling in early stages of development leads to the formation of gastruloids with anterior neural tissues. Thus, we provide a new tool to study the development of the mouse after implantation in vitro, especially the formation of anterior neural regions. Mouse embryonic stem cells can be aggregated to form epiblast-like (EPI) aggregates EPI aggregates undergo axial morphogenesis in the absence of exogenous WNT activation Initial aggregate size is a major determinant for axial morphogenesis Early WNT inhibition is essential for the emergence of anterior neural progenitors
Collapse
|
15
|
Chan CSY, Lonfat N, Zhao R, Davis AE, Li L, Wu MR, Lin CH, Ji Z, Cepko CL, Wang S. Cell type- and stage-specific expression of Otx2 is regulated by multiple transcription factors and cis-regulatory modules in the retina. Development 2020; 147:dev187922. [PMID: 32631829 PMCID: PMC7406324 DOI: 10.1242/dev.187922] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 06/22/2020] [Indexed: 12/22/2022]
Abstract
Transcription factors (TFs) are often used repeatedly during development and homeostasis to control distinct processes in the same and/or different cellular contexts. Considering the limited number of TFs in the genome and the tremendous number of events that need to be regulated, re-use of TFs is necessary. We analyzed how the expression of the homeobox TF, orthodenticle homeobox 2 (Otx2), is regulated in a cell type- and stage-specific manner during development in the mouse retina. We identified seven Otx2 cis-regulatory modules (CRMs), among which the O5, O7 and O9 CRMs mark three distinct cellular contexts of Otx2 expression. We discovered that Otx2, Crx and Sox2, which are well-known TFs regulating retinal development, bind to and activate the O5, O7 or O9 CRMs, respectively. The chromatin status of these three CRMs was found to be distinct in vivo in different retinal cell types and at different stages. We conclude that retinal cells use a cohort of TFs with different expression patterns and multiple CRMs with different chromatin configurations to regulate the expression of Otx2 precisely.
Collapse
Affiliation(s)
- Candace S Y Chan
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA 94304, USA
| | - Nicolas Lonfat
- Departments of Genetics and Ophthalmology, Blavatnik Institute, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Rong Zhao
- Departments of Genetics and Ophthalmology, Blavatnik Institute, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Alexander E Davis
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA 94304, USA
| | - Liang Li
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA 94304, USA
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Man-Ru Wu
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA 94304, USA
| | - Cheng-Hui Lin
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA 94304, USA
| | - Zhe Ji
- Department of Bioengineering, Northwestern University, Evanston, IL 60208, USA
| | - Constance L Cepko
- Departments of Genetics and Ophthalmology, Blavatnik Institute, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Sui Wang
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA 94304, USA
| |
Collapse
|
16
|
Vyas B, Nandkishore N, Sambasivan R. Vertebrate cranial mesoderm: developmental trajectory and evolutionary origin. Cell Mol Life Sci 2020; 77:1933-1945. [PMID: 31722070 PMCID: PMC11105048 DOI: 10.1007/s00018-019-03373-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/29/2019] [Accepted: 11/05/2019] [Indexed: 02/06/2023]
Abstract
Vertebrate cranial mesoderm is a discrete developmental unit compared to the mesoderm below the developing neck. An extraordinary feature of the cranial mesoderm is that it includes a common progenitor pool contributing to the chambered heart and the craniofacial skeletal muscles. This striking developmental potential and the excitement it generated led to advances in our understanding of cranial mesoderm developmental mechanism. Remarkably, recent findings have begun to unravel the origin of its distinct developmental characteristics. Here, we take a detailed view of the ontogenetic trajectory of cranial mesoderm and its regulatory network. Based on the emerging evidence, we propose that cranial and posterior mesoderm diverge at the earliest step of the process that patterns the mesoderm germ layer along the anterior-posterior body axis. Further, we discuss the latest evidence and their impact on our current understanding of the evolutionary origin of cranial mesoderm. Overall, the review highlights the findings from contemporary research, which lays the foundation to probe the molecular basis of unique developmental potential and evolutionary origin of cranial mesoderm.
Collapse
Affiliation(s)
- Bhakti Vyas
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bellary Road, Bengaluru, 560065, India
- Manipal Academy of Higher Education, Manipal, 576104, India
| | - Nitya Nandkishore
- Institute for Stem Cell Biology and Regenerative Medicine, GKVK Campus, Bellary Road, Bengaluru, 560065, India
- SASTRA University, Thirumalaisamudram, Thanjavur, 613401, India
| | - Ramkumar Sambasivan
- Indian Institute of Science Education and Research (IISER) Tirupati, Transit Campus, Karakambadi Road, Rami Reddy Nagar, Mangalam, Tirupati, Andhra Pradesh, 517507, India.
| |
Collapse
|
17
|
Raffaelli A, Stern CD. Signaling events regulating embryonic polarity and formation of the primitive streak in the chick embryo. Curr Top Dev Biol 2019; 136:85-111. [PMID: 31959299 DOI: 10.1016/bs.ctdb.2019.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The avian embryo is a key experimental model system for early development of amniotes. One key difference with invertebrates and "lower" vertebrates like fish and amphibians is that amniotes do not rely so heavily on maternal messages because the zygotic genome is activated very early. Early development also involves considerable growth in volume and mass of the embryo, with cell cycles that include G1 and G2 phases from very early cleavage. The very early maternal to zygotic transition also allows the embryo to establish its own polarity without relying heavily on maternal determinants. In many amniotes including avians and non-rodent mammals, this enables an ability of the embryo to "regulate": a single multicellular embryo can give rise to more than one individual-monozygotic twins. Here we discuss the embryological, cellular, molecular and evolutionary underpinnings of gastrulation in avian embryos as a model amniote embryo. Many of these properties are shared by human embryos.
Collapse
Affiliation(s)
- Ana Raffaelli
- Department of Cell & Developmental Biology, University College London, London, United Kingdom
| | - Claudio D Stern
- Department of Cell & Developmental Biology, University College London, London, United Kingdom.
| |
Collapse
|
18
|
McMahon R, Sibbritt T, Salehin N, Osteil P, Tam PPL. Mechanistic insights from the LHX1-driven molecular network in building the embryonic head. Dev Growth Differ 2019; 61:327-336. [PMID: 31111476 DOI: 10.1111/dgd.12609] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/05/2019] [Accepted: 04/08/2019] [Indexed: 12/27/2022]
Abstract
Development of an embryo is driven by a series of molecular instructions that control the differentiation of tissue precursor cells and shape the tissues into major body parts. LIM homeobox 1 (LHX1) is a transcription factor that plays a major role in the development of the embryonic head of the mouse. Loss of LHX1 function disrupts the morphogenetic movement of head tissue precursors and impacts on the function of molecular factors in modulating the activity of the WNT signaling pathway. LHX1 acts with a transcription factor complex to regulate the transcription of target genes in multiple phases of development and in a range of embryonic tissues of the mouse and Xenopus. Determining the interacting factors and transcriptional targets of LHX1 will be key to unraveling the ensemble of factors involved in head development and building a head gene regulatory network.
Collapse
Affiliation(s)
- Riley McMahon
- Embryology Unit, Children's Medical Research Institute, The University of Sydney, Westmead, New South Wales, Australia.,Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Westmead, New South Wales, Australia
| | - Tennille Sibbritt
- Embryology Unit, Children's Medical Research Institute, The University of Sydney, Westmead, New South Wales, Australia.,Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Westmead, New South Wales, Australia
| | - Nazmus Salehin
- Embryology Unit, Children's Medical Research Institute, The University of Sydney, Westmead, New South Wales, Australia.,Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Westmead, New South Wales, Australia
| | - Pierre Osteil
- Embryology Unit, Children's Medical Research Institute, The University of Sydney, Westmead, New South Wales, Australia.,Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Westmead, New South Wales, Australia
| | - Patrick P L Tam
- Embryology Unit, Children's Medical Research Institute, The University of Sydney, Westmead, New South Wales, Australia.,Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Westmead, New South Wales, Australia
| |
Collapse
|
19
|
Zhang S, Chen T, Chen N, Gao D, Shi B, Kong S, West RC, Yuan Y, Zhi M, Wei Q, Xiang J, Mu H, Yue L, Lei X, Wang X, Zhong L, Liang H, Cao S, Belmonte JCI, Wang H, Han J. Implantation initiation of self-assembled embryo-like structures generated using three types of mouse blastocyst-derived stem cells. Nat Commun 2019; 10:496. [PMID: 30700702 PMCID: PMC6353907 DOI: 10.1038/s41467-019-08378-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 01/04/2019] [Indexed: 11/09/2022] Open
Abstract
Spatially ordered embryo-like structures self-assembled from blastocyst-derived stem cells can be generated to mimic embryogenesis in vitro. However, the assembly system and developmental potential of such structures needs to be further studied. Here, we devise a nonadherent-suspension-shaking system to generate self-assembled embryo-like structures (ETX-embryoids) using mouse embryonic, trophoblast and extra-embryonic endoderm stem cells. When cultured together, the three cell types aggregate and sort into lineage-specific compartments. Signaling among these compartments results in molecular and morphogenic events that closely mimic those observed in wild-type embryos. These ETX-embryoids exhibit lumenogenesis, asymmetric patterns of gene expression for markers of mesoderm and primordial germ cell precursors, and formation of anterior visceral endoderm-like tissues. After transplantation into the pseudopregnant mouse uterus, ETX-embryoids efficiently initiate implantation and trigger the formation of decidual tissues. The ability of the three cell types to self-assemble into an embryo-like structure in vitro provides a powerful model system for studying embryogenesis.
Collapse
Affiliation(s)
- Shaopeng Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 10094, China
| | - Tianzhi Chen
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 10094, China
| | - Naixin Chen
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 10094, China
| | - Dengfeng Gao
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 10094, China
| | - Bingbo Shi
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 10094, China
| | - Shuangbo Kong
- Fujian Provincial Key Laboratory of Reproductive Health Research, Medical College of Xiamen University, Xiamen, Fujian, 361102, China
| | | | - Ye Yuan
- Colorado Center for Reproductive Medicine, Lone Tree, CO, 80124, USA
| | - Minglei Zhi
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 10094, China
| | - Qingqing Wei
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 10094, China
| | - Jinzhu Xiang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 10094, China
| | - Haiyuan Mu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 10094, China
| | - Liang Yue
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 10094, China
| | - Xiaohua Lei
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xuepeng Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Liang Zhong
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Hui Liang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 10094, China
| | - Suying Cao
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, 102206, China
| | | | - Haibin Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Medical College of Xiamen University, Xiamen, Fujian, 361102, China
| | - Jianyong Han
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 10094, China. .,Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, 100083, China.
| |
Collapse
|
20
|
Sibbritt T, Ip CK, Khoo P, Wilkie E, Jones V, Sun JQJ, Shen JX, Peng G, Han JJ, Jing N, Osteil P, Ramialison M, Tam PPL, Fossat N. A gene regulatory network anchored by LIM homeobox 1 for embryonic head development. Genesis 2018; 56:e23246. [DOI: 10.1002/dvg.23246] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/09/2018] [Accepted: 08/09/2018] [Indexed: 02/02/2023]
Affiliation(s)
- Tennille Sibbritt
- Embryology Unit, Children's Medical Research Institute The University of Sydney Sydney New South Wales Australia
- School of Medical Sciences, Faculty of Medicine and Health The University of Sydney Sydney New South Wales Australia
| | - Chi K. Ip
- Embryology Unit, Children's Medical Research Institute The University of Sydney Sydney New South Wales Australia
| | - Poh‐Lynn Khoo
- Embryology Unit, Children's Medical Research Institute The University of Sydney Sydney New South Wales Australia
| | - Emilie Wilkie
- Embryology Unit, Children's Medical Research Institute The University of Sydney Sydney New South Wales Australia
- Bioinformatics Unit, Children's Medical Research Institute The University of Sydney Sydney New South Wales Australia
| | - Vanessa Jones
- Embryology Unit, Children's Medical Research Institute The University of Sydney Sydney New South Wales Australia
| | - Jane Q. J. Sun
- Embryology Unit, Children's Medical Research Institute The University of Sydney Sydney New South Wales Australia
| | - Joanne X. Shen
- Embryology Unit, Children's Medical Research Institute The University of Sydney Sydney New South Wales Australia
| | - Guangdun Peng
- State Key Laboratory of Cell Biology Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences Shanghai China
| | - Jing‐Dong J. Han
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences‐Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences Chinese Academy of Sciences Shanghai China
| | - Naihe Jing
- State Key Laboratory of Cell Biology Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences Shanghai China
- School of Life Science and Technology ShanghaiTech University Shanghai China
| | - Pierre Osteil
- Embryology Unit, Children's Medical Research Institute The University of Sydney Sydney New South Wales Australia
- School of Medical Sciences, Faculty of Medicine and Health The University of Sydney Sydney New South Wales Australia
| | - Mirana Ramialison
- Australian Regenerative Medicine Institute Monash University Melbourne Victoria Australia
| | - Patrick P. L. Tam
- Embryology Unit, Children's Medical Research Institute The University of Sydney Sydney New South Wales Australia
- School of Medical Sciences, Faculty of Medicine and Health The University of Sydney Sydney New South Wales Australia
| | - Nicolas Fossat
- Embryology Unit, Children's Medical Research Institute The University of Sydney Sydney New South Wales Australia
- School of Medical Sciences, Faculty of Medicine and Health The University of Sydney Sydney New South Wales Australia
| |
Collapse
|
21
|
Abstract
We present an overview of symmetry breaking in early mammalian development as a continuous process from compaction to specification of the body axes. While earlier studies have focused on individual symmetry-breaking events, recent advances enable us to explore progressive symmetry breaking during early mammalian development. Although we primarily discuss embryonic development of the mouse, as it is the best-studied mammalian model system to date, we also highlight the shared and distinct aspects between different mammalian species. Finally, we discuss how insights gained from studying mammalian development can be generalized in light of self-organization principles. With this review, we hope to highlight new perspectives in studying symmetry breaking and self-organization in multicellular systems.
Collapse
Affiliation(s)
- Hui Ting Zhang
- European Molecular Biology Laboratory, 69117 Heidelberg, Germany;
| | - Takashi Hiiragi
- European Molecular Biology Laboratory, 69117 Heidelberg, Germany;
| |
Collapse
|
22
|
Abstract
TGF-β family ligands function in inducing and patterning many tissues of the early vertebrate embryonic body plan. Nodal signaling is essential for the specification of mesendodermal tissues and the concurrent cellular movements of gastrulation. Bone morphogenetic protein (BMP) signaling patterns tissues along the dorsal-ventral axis and simultaneously directs the cell movements of convergence and extension. After gastrulation, a second wave of Nodal signaling breaks the symmetry between the left and right sides of the embryo. During these processes, elaborate regulatory feedback between TGF-β ligands and their antagonists direct the proper specification and patterning of embryonic tissues. In this review, we summarize the current knowledge of the function and regulation of TGF-β family signaling in these processes. Although we cover principles that are involved in the development of all vertebrate embryos, we focus specifically on three popular model organisms: the mouse Mus musculus, the African clawed frog of the genus Xenopus, and the zebrafish Danio rerio, highlighting the similarities and differences between these species.
Collapse
Affiliation(s)
- Joseph Zinski
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104-6058
| | - Benjamin Tajer
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104-6058
| | - Mary C Mullins
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104-6058
| |
Collapse
|
23
|
Stokes BA, Sabatino JA, Zohn IE. High levels of iron supplementation prevents neural tube defects in the Fpn1 ffe mouse model. Birth Defects Res 2018; 109:81-91. [PMID: 28008752 DOI: 10.1002/bdra.23542] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 05/11/2016] [Accepted: 06/05/2016] [Indexed: 12/16/2022]
Abstract
BACKGROUND Periconception maternal nutrition and folate in particular are important factors influencing the incidence of neural tube defects (NTDs). Many but not all NTDs are prevented by folic acid supplementation and there is a pressing need for additional strategies to prevent these birth defects. Other micronutrients such as iron are potential candidates, yet a clear role for iron deficiency in contributing to NTDs is lacking. Our previous studies with the flatiron (ffe) mouse model of Ferroportin1 (Fpn1) deficiency suggest that iron is required for neural tube closure and forebrain development raising the possibility that iron supplementation could prevent NTDs. METHODS We determined the effect of periconception iron and/or folic acid supplementation on the penetrance of NTDs in the Fpn1ffe mouse model. Concurrently, measurements of folate and iron were made to ensure supplementation had the intended effects. RESULTS High levels of iron supplementation significantly reduced the incidence of NTDs in Fpn1ffe mutants. Fpn1 deficiency resulted in reduced folate levels in both pregnant dams and embryos. Yet folic acid supplementation did not prevent NTDs in the Fpn1ffe model. Similarly, forebrain truncations were rescued with iron. Surprisingly, the high levels of iron supplementation used in this study caused folate deficiency in wild-type dams and embryos. CONCLUSION Our results demonstrate that iron supplementation can prevent NTDs and forebrain truncations in the Fpn1ffe model. Surprisingly, high levels of iron supplementation and iron overload can cause folate deficiency. If iron is essential for neural tube closure, it is possible that iron deficiency might contribute to NTDs. Birth Defects Research 109:81-91, 2017. © 2016 The Authors Birth Defects Research Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Bethany A Stokes
- Department of Biology, The George Washington University, Washington, DC.,Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC
| | - Julia A Sabatino
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC
| | - Irene E Zohn
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC
| |
Collapse
|
24
|
Stower MJ, Srinivas S. The Head's Tale: Anterior-Posterior Axis Formation in the Mouse Embryo. Curr Top Dev Biol 2017; 128:365-390. [PMID: 29477169 DOI: 10.1016/bs.ctdb.2017.11.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The establishment of the anterior-posterior (A-P) axis is a fundamental event during early development and marks the start of the process by which the basic body plan is laid down. This axial information determines where gastrulation, that generates and positions cells of the three-germ layers, occurs. A-P patterning requires coordinated interactions between multiple tissues, tight spatiotemporal control of signaling pathways, and the coordination of tissue growth with morphogenetic movements. In the mouse, a specialized population of cells, the anterior visceral endoderm (AVE) undergoes a migration event critical for correct A-P pattern. In this review, we summarize our understanding of the generation of anterior pattern, focusing on the role of the AVE. We will also outline some of the many questions that remain regarding the mechanism by which the first axial asymmetry is established, how the AVE is induced, and how it moves within the visceral endoderm epithelium.
Collapse
|
25
|
Home P, Kumar RP, Ganguly A, Saha B, Milano-Foster J, Bhattacharya B, Ray S, Gunewardena S, Paul A, Camper SA, Fields PE, Paul S. Genetic redundancy of GATA factors in the extraembryonic trophoblast lineage ensures the progression of preimplantation and postimplantation mammalian development. Development 2017; 144:876-888. [PMID: 28232602 PMCID: PMC5374352 DOI: 10.1242/dev.145318] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 01/10/2017] [Indexed: 12/16/2022]
Abstract
GATA transcription factors are implicated in establishing cell fate during mammalian development. In early mammalian embryos, GATA3 is selectively expressed in the extraembryonic trophoblast lineage and regulates gene expression to promote trophoblast fate. However, trophoblast-specific GATA3 function is dispensable for early mammalian development. Here, using dual conditional knockout mice, we show that genetic redundancy of Gata3 with paralog Gata2 in trophoblast progenitors ensures the successful progression of both pre- and postimplantation mammalian development. Stage-specific gene deletion in trophoblasts reveals that loss of both GATA genes, but not either alone, leads to embryonic lethality prior to the onset of their expression within the embryo proper. Using ChIP-seq and RNA-seq analyses, we define the global targets of GATA2/GATA3 and show that they directly regulate a large number of common genes to orchestrate stem versus differentiated trophoblast fate. In trophoblast progenitors, GATA factors directly regulate BMP4, Nodal and Wnt signaling components that promote embryonic-extraembryonic signaling cross-talk, which is essential for the development of the embryo proper. Our study provides genetic evidence that impairment of trophoblast-specific GATA2/GATA3 function could lead to early pregnancy failure. Summary: During trophoblast development in mice, GATA2 and GATA3 act synergistically by directly regulating a large number of common genes, and together are important to ensure trophoblast lineage progression.
Collapse
Affiliation(s)
- Pratik Home
- Department of Pathology and Laboratory Medicine and Institute for Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Ram Parikshan Kumar
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Avishek Ganguly
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Biswarup Saha
- Department of Pathology and Laboratory Medicine and Institute for Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Jessica Milano-Foster
- Department of Pathology and Laboratory Medicine and Institute for Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Bhaswati Bhattacharya
- Department of Pathology and Laboratory Medicine and Institute for Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Soma Ray
- Department of Pathology and Laboratory Medicine and Institute for Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Sumedha Gunewardena
- Department of Molecular and Integrative Physiology, Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Arindam Paul
- Department of Pathology and Laboratory Medicine and Institute for Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Sally A Camper
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Patrick E Fields
- Department of Pathology and Laboratory Medicine and Institute for Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Soumen Paul
- Department of Pathology and Laboratory Medicine and Institute for Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
26
|
Shioi G, Hoshino H, Abe T, Kiyonari H, Nakao K, Meng W, Furuta Y, Fujimori T, Aizawa S. Apical constriction in distal visceral endoderm cells initiates global, collective cell rearrangement in embryonic visceral endoderm to form anterior visceral endoderm. Dev Biol 2017; 429:20-30. [PMID: 28712875 DOI: 10.1016/j.ydbio.2017.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 07/10/2017] [Accepted: 07/10/2017] [Indexed: 12/11/2022]
Abstract
The behavior of visceral endoderm cells was examined as the anterior visceral endoderm (AVE) formed from the distal visceral endoderm (DVE) using the mouse lines R26-H2B-EGFP and R26-PHA7-EGFP to visualize cell nuclei and adherens junction, respectively. The analysis using R26-H2B-EGFP demonstrated global cell rearrangement that was not specific to the DVE cells in the monolayer embryonic visceral endoderm sheet; each population of the endoderm cells moved collectively in a swirling movement as a whole. Most of the AVE cells at E6.5 were not E5.5 DVE cells but were E5.5 cells that were located caudally behind them, as previously reported (Hoshino et al., 2015; Takaoka et al., 2011). In the rearrangement, the posterior embryonic visceral endoderm cells did not move, as extraembryonic visceral endoderm cells did not, and they constituted a distinct population during the process of anterior-posterior axis formation. The analysis using R26-PHA7-EGFP suggested that constriction of the apical surfaces of the cells in prospective anterior portion of the DVE initiated the global cellular movement of the embryonic visceral endoderm to drive AVE formation.
Collapse
Affiliation(s)
- Go Shioi
- Genetic Engineering Team, RIKEN Center for Life Science Technologies (CLST), 2-2-3 Minatojima Minami-machi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.
| | - Hideharu Hoshino
- Laboratory for Vertebrate Body Plan, RIKEN Center for Developmental Biology (CDB), 2-2-3 Minatojima Minami-machi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Takaya Abe
- Genetic Engineering Team, RIKEN Center for Life Science Technologies (CLST), 2-2-3 Minatojima Minami-machi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Hiroshi Kiyonari
- Genetic Engineering Team, RIKEN Center for Life Science Technologies (CLST), 2-2-3 Minatojima Minami-machi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Animal Resource Development Unit, RIKEN Center for Life Science Technologies (CLST), 2-2-3 Minatojima Minami-machi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Kazuki Nakao
- Animal Resource Development Unit, RIKEN Center for Life Science Technologies (CLST), 2-2-3 Minatojima Minami-machi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine (CDBIM), University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Wenxiang Meng
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Yasuhide Furuta
- Genetic Engineering Team, RIKEN Center for Life Science Technologies (CLST), 2-2-3 Minatojima Minami-machi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Animal Resource Development Unit, RIKEN Center for Life Science Technologies (CLST), 2-2-3 Minatojima Minami-machi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Toshihiko Fujimori
- Genetic Engineering Team, RIKEN Center for Life Science Technologies (CLST), 2-2-3 Minatojima Minami-machi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Division of Embryology, National Institute for Basic Biology (NIBB), 5-1 Higashiyama, Myodaiji-cho, Okazaki, Aichi 444-8787, Japan
| | - Shinichi Aizawa
- Genetic Engineering Team, RIKEN Center for Life Science Technologies (CLST), 2-2-3 Minatojima Minami-machi, Chuo-ku, Kobe, Hyogo 650-0047, Japan; Laboratory for Vertebrate Body Plan, RIKEN Center for Developmental Biology (CDB), 2-2-3 Minatojima Minami-machi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| |
Collapse
|
27
|
Amirpour N, Razavi S, Esfandiari E, Hashemibeni B, Kazemi M, Salehi H. Hanging drop culture enhances differentiation of human adipose-derived stem cells into anterior neuroectodermal cells using small molecules. Int J Dev Neurosci 2017; 59:21-30. [PMID: 28285945 DOI: 10.1016/j.ijdevneu.2017.03.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 03/04/2017] [Accepted: 03/05/2017] [Indexed: 01/26/2023] Open
Abstract
Inspired by in vivo developmental process, several studies were conducted to design a protocol for differentiating of mesenchymal stem cells into neural cells in vitro. Human adipose-derived stem cells (hADSCs) as mesenchymal stem cells are a promising source for this purpose. At current study, we applied a defined neural induction medium by using small molecules for direct differentiation of hADSCs into anterior neuroectodermal cells. Anterior neuroectodermal differentiation of hADSCs was performed by hanging drop and monolayer protocols. At these methods, three small molecules were used to suppress the BMP, Nodal, and Wnt signaling pathways in order to obtain anterior neuroectodermal (eye field) cells from hADSCs. After two and three weeks of induction, the differentiated cells with neural morphology expressed anterior neuroectodermal markers such as OTX2, SIX3, β-TUB III and PAX6. The protein expression of such markers was confirmed by real time, RT-PCR and immunocytochemistry methods According to our data, it seems that the hanging drop method is a proper approach for neuroectodermal induction of hADSCs. Considering wide availability and immunosuppressive properties of hADSCs, these cells may open a way for autologous cell therapy of neurodegenerative disorders.
Collapse
Affiliation(s)
- Noushin Amirpour
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shahnaz Razavi
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ebrahim Esfandiari
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Batoul Hashemibeni
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Kazemi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Salehi
- Department of Anatomical Sciences and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
28
|
Matsuo I, Hiramatsu R. Mechanical perspectives on the anterior-posterior axis polarization of mouse implanted embryos. Mech Dev 2017; 144:62-70. [DOI: 10.1016/j.mod.2016.09.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 09/20/2016] [Accepted: 09/29/2016] [Indexed: 01/21/2023]
|
29
|
Houston DW. Vertebrate Axial Patterning: From Egg to Asymmetry. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 953:209-306. [PMID: 27975274 PMCID: PMC6550305 DOI: 10.1007/978-3-319-46095-6_6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The emergence of the bilateral embryonic body axis from a symmetrical egg has been a long-standing question in developmental biology. Historical and modern experiments point to an initial symmetry-breaking event leading to localized Wnt and Nodal growth factor signaling and subsequent induction and formation of a self-regulating dorsal "organizer." This organizer forms at the site of notochord cell internalization and expresses primarily Bone Morphogenetic Protein (BMP) growth factor antagonists that establish a spatiotemporal gradient of BMP signaling across the embryo, directing initial cell differentiation and morphogenesis. Although the basics of this model have been known for some time, many of the molecular and cellular details have only recently been elucidated and the extent that these events remain conserved throughout vertebrate evolution remains unclear. This chapter summarizes historical perspectives as well as recent molecular and genetic advances regarding: (1) the mechanisms that regulate symmetry-breaking in the vertebrate egg and early embryo, (2) the pathways that are activated by these events, in particular the Wnt pathway, and the role of these pathways in the formation and function of the organizer, and (3) how these pathways also mediate anteroposterior patterning and axial morphogenesis. Emphasis is placed on comparative aspects of the egg-to-embryo transition across vertebrates and their evolution. The future prospects for work regarding self-organization and gene regulatory networks in the context of early axis formation are also discussed.
Collapse
Affiliation(s)
- Douglas W Houston
- Department of Biology, The University of Iowa, 257 BB, Iowa City, IA, 52242, USA.
| |
Collapse
|
30
|
Conserved and divergent expression patterns of markers of axial development in the laboratory opossum,Monodelphis domestica. Dev Dyn 2016; 245:1176-1188. [DOI: 10.1002/dvdy.24459] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 09/14/2016] [Accepted: 09/14/2016] [Indexed: 11/07/2022] Open
|
31
|
Epiblast-specific loss of HCF-1 leads to failure in anterior-posterior axis specification. Dev Biol 2016; 418:75-88. [PMID: 27521049 DOI: 10.1016/j.ydbio.2016.08.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/08/2016] [Accepted: 08/08/2016] [Indexed: 02/06/2023]
Abstract
Mammalian Host-Cell Factor 1 (HCF-1), a transcriptional co-regulator, plays important roles during the cell-division cycle in cell culture, embryogenesis as well as adult tissue. In mice, HCF-1 is encoded by the X-chromosome-linked Hcfc1 gene. Induced Hcfc1(cKO/+) heterozygosity with a conditional knockout (cKO) allele in the epiblast of female embryos leads to a mixture of HCF-1-positive and -deficient cells owing to random X-chromosome inactivation. These embryos survive owing to the replacement of all HCF-1-deficient cells by HCF-1-positive cells during E5.5 to E8.5 of development. In contrast, complete epiblast-specific loss of HCF-1 in male embryos, Hcfc1(epiKO/Y), leads to embryonic lethality. Here, we characterize this lethality. We show that male epiblast-specific loss of Hcfc1 leads to a developmental arrest at E6.5 with a rapid progressive cell-cycle exit and an associated failure of anterior visceral endoderm migration and primitive streak formation. Subsequently, gastrulation does not take place. We note that the pattern of Hcfc1(epiKO/Y) lethality displays many similarities to loss of β-catenin function. These results reveal essential new roles for HCF-1 in early embryonic cell proliferation and development.
Collapse
|
32
|
Yoshida M, Kajikawa E, Kurokawa D, Noro M, Iwai T, Yonemura S, Kobayashi K, Kiyonari H, Aizawa S. Conserved and divergent expression patterns of markers of axial development in reptilian embryos: Chinese soft-shell turtle and Madagascar ground gecko. Dev Biol 2016; 415:122-142. [DOI: 10.1016/j.ydbio.2016.05.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 05/06/2016] [Accepted: 05/06/2016] [Indexed: 12/18/2022]
|
33
|
Henrique D, Abranches E, Verrier L, Storey KG. Neuromesodermal progenitors and the making of the spinal cord. Development 2015; 142:2864-75. [PMID: 26329597 PMCID: PMC4958456 DOI: 10.1242/dev.119768] [Citation(s) in RCA: 230] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Neuromesodermal progenitors (NMps) contribute to both the elongating spinal cord and the adjacent paraxial mesoderm. It has been assumed that these cells arise as a result of patterning of the anterior neural plate. However, as the molecular mechanisms that specify NMps in vivo are uncovered, and as protocols for generating these bipotent cells from mouse and human pluripotent stem cells in vitro are established, the emerging data suggest that this view needs to be revised. Here, we review the characteristics, regulation, in vitro derivation and in vivo induction of NMps. We propose that these cells arise within primitive streak-associated epiblast via a mechanism that is separable from that which establishes neural fate in the anterior epiblast. We thus argue for the existence of two distinct routes for making central nervous system progenitors.
Collapse
Affiliation(s)
- Domingos Henrique
- Instituto de Medicina Molecular and Instituto de Histologia e Biologia do Desenvolvimento, Faculdade de Medicina da Universidade de Lisboa, Avenida Prof. Egas Moniz, Lisboa 1649-028, Portugal
| | - Elsa Abranches
- Instituto de Medicina Molecular and Instituto de Histologia e Biologia do Desenvolvimento, Faculdade de Medicina da Universidade de Lisboa, Avenida Prof. Egas Moniz, Lisboa 1649-028, Portugal
| | - Laure Verrier
- Division of Cell & Developmental Biology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| | - Kate G Storey
- Division of Cell & Developmental Biology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| |
Collapse
|
34
|
Yoshida M, Kajikawa E, Kurokawa D, Tokunaga T, Onishi A, Yonemura S, Kobayashi K, Kiyonari H, Aizawa S. Conserved and divergent expression patterns of markers of axial development in eutherian mammals. Dev Dyn 2015; 245:67-86. [PMID: 26404161 DOI: 10.1002/dvdy.24352] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 09/12/2015] [Accepted: 09/12/2015] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Mouse embryos are cup shaped, but most nonrodent eutherian embryos are disk shaped. Extraembryonic ectoderm (ExEc), which may have essential roles in anterior-posterior (A-P) axis formation in mouse embryos, does not develop in many eutherian embryos. To assess A-P axis formation in eutherians, comparative analyses were made on rabbit, porcine, and Suncus embryos. RESULTS All embryos examined expressed Nodal initially throughout epiblast and visceral endoderm; its expression became restricted to the posterior region before gastrulation. Anterior visceral endoderm (AVE) genes were expressed in Otx2-positive visceral endoderm, with Dkk1 expression being most anterior. The mouse pattern of AVE formation was conserved in rabbit embryos, but had diverged in porcine and Suncus embryos. No structure that was molecularly equivalent to Bmp-positive ExEc, existed in rabbit or pig embryos. In Suncus embryos, A-P axis was determined at prehatching stage, and these embryos attached to uterine wall at future posterior side. CONCLUSIONS Nodal, but not Bmp, functions in epiblast and visceral endoderm development may be conserved in eutherians. AVE functions may also be conserved, but the pattern of its formation has diverged among eutherians. Roles of BMP and NODAL gradients in AVE formation seem to have been established in a subset of rodents.
Collapse
Affiliation(s)
- Michio Yoshida
- Laboratory for Vertebrate Body Plan, Center for Developmental Biology (CDB), RIKEN Kobe, Chuo-ku, Kobe, Japan
| | - Eriko Kajikawa
- Laboratory for Vertebrate Body Plan, Center for Developmental Biology (CDB), RIKEN Kobe, Chuo-ku, Kobe, Japan
| | - Daisuke Kurokawa
- Laboratory for Vertebrate Body Plan, Center for Developmental Biology (CDB), RIKEN Kobe, Chuo-ku, Kobe, Japan.,Misaki Marine Biological Station, Graduate School of Science, The University of Tokyo, Misaki, Miura, Kanagawa, Japan
| | - Tomoyuki Tokunaga
- Animal Development and Differentiation Research Unit, Animal Research Division, National Institute of Agrobiological Sciences (NIAS), Tsukuba-shi, Ibaraki, Japan
| | - Akira Onishi
- Laboratory of Animal Reproduction, Department of Animal Science and Resources, Nihon University College of Bioresource Sciences, Fujisawa, Kanagawa, Japan
| | - Shigenobu Yonemura
- Ultrastructural Research Team, Biosystem Dynamics Group, Division of Bio-function Dynamics Imaging, RIKEN Center for Life Science Technologies (CLST), Chuo-ku, Kobe, Japan
| | - Kensaku Kobayashi
- Laboratory for Animal Resources and Genetic Engineering, Center for Developmental Biology (CDB), RIKEN Kobe, Chuo-ku, Kobe, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, Center for Developmental Biology (CDB), RIKEN Kobe, Chuo-ku, Kobe, Japan
| | - Shinichi Aizawa
- Laboratory for Vertebrate Body Plan, Center for Developmental Biology (CDB), RIKEN Kobe, Chuo-ku, Kobe, Japan.,Laboratory for Animal Resources and Genetic Engineering, Center for Developmental Biology (CDB), RIKEN Kobe, Chuo-ku, Kobe, Japan
| |
Collapse
|
35
|
Parfitt DE, Shen MM. From blastocyst to gastrula: gene regulatory networks of embryonic stem cells and early mouse embryogenesis. Philos Trans R Soc Lond B Biol Sci 2015; 369:rstb.2013.0542. [PMID: 25349451 DOI: 10.1098/rstb.2013.0542] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
To date, many regulatory genes and signalling events coordinating mammalian development from blastocyst to gastrulation stages have been identified by mutational analyses and reverse-genetic approaches, typically on a gene-by-gene basis. More recent studies have applied bioinformatic approaches to generate regulatory network models of gene interactions on a genome-wide scale. Such models have provided insights into the gene networks regulating pluripotency in embryonic and epiblast stem cells, as well as cell-lineage determination in vivo. Here, we review how regulatory networks constructed for different stem cell types relate to corresponding networks in vivo and provide insights into understanding the molecular regulation of the blastocyst-gastrula transition.
Collapse
Affiliation(s)
- David-Emlyn Parfitt
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA Department of Genetics and Development, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA Department of Urology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA Department of Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Michael M Shen
- Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA Department of Genetics and Development, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA Department of Urology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA Department of Systems Biology, Herbert Irving Comprehensive Cancer Center, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| |
Collapse
|
36
|
Stower MJ, Srinivas S. Heading forwards: anterior visceral endoderm migration in patterning the mouse embryo. Philos Trans R Soc Lond B Biol Sci 2015; 369:rstb.2013.0546. [PMID: 25349454 PMCID: PMC4216468 DOI: 10.1098/rstb.2013.0546] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The elaboration of anterior–posterior (A–P) pattern is one of the earliest events during development and requires the precisely coordinated action of several players at the level of molecules, cells and tissues. In mammals, it is controlled by a specialized population of migratory extraembryonic epithelial cells, the anterior visceral endoderm (AVE). The AVE is a signalling centre that is responsible for several important patterning events during early development, including specifying the orientation of the A–P axis and the position of the heart with respect to the brain. AVE cells undergo a characteristic stereotypical migration which is crucial to their functions.
Collapse
Affiliation(s)
- Matthew J Stower
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Shankar Srinivas
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| |
Collapse
|
37
|
Hoshino H, Shioi G, Aizawa S. AVE protein expression and visceral endoderm cell behavior during anterior-posterior axis formation in mouse embryos: Asymmetry in OTX2 and DKK1 expression. Dev Biol 2015; 402:175-91. [PMID: 25910836 DOI: 10.1016/j.ydbio.2015.03.023] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 03/20/2015] [Accepted: 03/21/2015] [Indexed: 11/27/2022]
Abstract
The initial landmark of anterior-posterior (A-P) axis formation in mouse embryos is the distal visceral endoderm, DVE, which expresses a series of anterior genes at embryonic day 5.5 (E5.5). Subsequently, DVE cells move to the future anterior region, generating anterior visceral endoderm (AVE). Questions remain regarding how the DVE is formed and how the direction of the movement is determined. This study compares the detailed expression patterns of OTX2, HHEX, CER1, LEFTY1 and DKK1 by immunohistology and live imaging at E4.5-E6.5. At E6.5, the AVE is subdivided into four domains: most anterior (OTX2, HHEX, CER1-low/DKK1-high), anterior (OTX2, HHEX, CER1-high/DKK1-low), main (OTX2, HHEX, CER1, LEFTY1-high) and antero-lateral and posterior (OTX2, HHEX-low). The study demonstrates how this pattern is established. AVE protein expression in the DVE occurs de novo at E5.25-E5.5. Neither HHEX, LEFTY1 nor CER1 expression is asymmetric. In contrast, OTX2 expression is tilted on the future posterior side with the DKK1 expression at its proximal domain; the DVE cells move in the opposite direction of the tilt.
Collapse
Affiliation(s)
- Hideharu Hoshino
- Laboratory for Vertebrate Body Plan, Center for Developmental Biology (CDB), RIKEN Kobe, 2-2-3 Minatojima Minami-machi, Chuo-ku, Kobe 650-0046, Japan.
| | - Go Shioi
- Laboratory for Animal Resources and Genetic Engineering, Center for Developmental Biology (CDB), RIKEN Kobe, 2-2-3 Minatojima Minami-machi, Chuo-ku, Kobe 650-0046, Japan.
| | - Shinichi Aizawa
- Laboratory for Vertebrate Body Plan, Center for Developmental Biology (CDB), RIKEN Kobe, 2-2-3 Minatojima Minami-machi, Chuo-ku, Kobe 650-0046, Japan; Laboratory for Animal Resources and Genetic Engineering, Center for Developmental Biology (CDB), RIKEN Kobe, 2-2-3 Minatojima Minami-machi, Chuo-ku, Kobe 650-0046, Japan.
| |
Collapse
|
38
|
Li L, Song L, Liu C, Chen J, Peng G, Wang R, Liu P, Tang K, Rossant J, Jing N. Ectodermal progenitors derived from epiblast stem cells by inhibition of Nodal signaling. J Mol Cell Biol 2015; 7:455-65. [PMID: 25990320 DOI: 10.1093/jmcb/mjv030] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 01/27/2015] [Indexed: 12/14/2022] Open
Abstract
The ectoderm has the capability to generate epidermis and neuroectoderm and plays imperative roles during the early embryonic development. Our recent study uncovered a region with ectodermal progenitor potential in mouse embryo at embryonic day 7.0 and revealed that Nodal inhibition is essential for its formation. Here, we demonstrate that through brief inhibition of Nodal signaling in vitro, mouse embryonic stem cell (ESC)-derived epiblast stem cells (ESD-EpiSCs) could be committed to transient ectodermal progenitor populations, which possess the ability to give rise to neural or epidermal ectoderm in the absence or presence of BMP4, respectively. Mechanistic studies reveal that BMP4 recruits distinct transcriptional targets in ESD-EpiSCs and ectoderm-like cells. Furthermore, FGF-Erk signaling may also be alleviated during the generation of ectoderm-like cells. Thus, our data suggest that instructive interactions among several extracellular signals participate in the commitment of ectoderm from ESD-EpiSCs, which shed new light on the understanding of the formation of ectoderm during the gastrulation in early mouse embryo development.
Collapse
Affiliation(s)
- Lingyu Li
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China Present address: Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lu Song
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chang Liu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jun Chen
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Guangdun Peng
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ran Wang
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Pingyu Liu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ke Tang
- Institute of Life Science, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Janet Rossant
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children Research Institute, Toronto, ON M5G 1X8, Canada
| | - Naihe Jing
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
39
|
Cabral-Teixeira J, Martinez-Fernandez A, Cai W, Terzic A, Mercola M, Willems E. Cholesterol-derived glucocorticoids control early fate specification in embryonic stem cells. Stem Cell Res 2015; 15:88-95. [PMID: 26024790 DOI: 10.1016/j.scr.2015.04.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 04/25/2015] [Accepted: 04/27/2015] [Indexed: 10/23/2022] Open
Abstract
Aside from its role in cell membrane integrity, cholesterol is a key component in steroid hormone production. The vital functions of steroid hormones such as estrogen, testosterone, glucocorticoids (Gcrts) and mineralocorticoids (Mnrts) in perinatal and adult life are well understood; however, their role during early embryonic development remains largely unexplored. Here we show that siRNA-mediated perturbation of steroid hormone production during mesoderm formation has important consequences on cardiac differentiation in mouse embryonic stem cells (mESC). Both Gcrts and Mnrts are capable of driving cardiac differentiation in mESC. Interestingly, the Gcrt receptor is widely expressed during gastrulation in the mouse, and is exclusively localized in the nuclei-and thus active-in visceral endoderm cells, suggesting that it functions much earlier than previously anticipated. We therefore studied Gcrt signaling in mESC as a model of the gastrulating embryo, and found that Gcrt signaling regulates expression of the transcription factor Hnf4a and the secreted Nodal and BMP inhibitor Cer1 in the early visceral endoderm. RNAi-mediated knockdown of Gcrt function blocked cardiomyocyte differentiation, with limited effects on other cardiovascular cell types including vascular endothelial cells and smooth muscle. Furthermore, the cardiogenic effect of Gcrts required Hnf4a and paracrine Cer1. These results establish a novel function for cholesterol-derived steroid hormones and identify Gcrt signaling in visceral endoderm cells as a regulator of Cer1 and cardiac fate.
Collapse
Affiliation(s)
- Joaquim Cabral-Teixeira
- Muscle Development and Regeneration Program, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA; Graduate Program in Molecular Pathology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | | | - Wenqing Cai
- Muscle Development and Regeneration Program, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA; Graduate School of Biomedical Sciences, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - Andre Terzic
- Division of Cardiovascular Diseases and Center for Regenerative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Mark Mercola
- Muscle Development and Regeneration Program, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA; Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Erik Willems
- Muscle Development and Regeneration Program, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA.
| |
Collapse
|
40
|
Meganathan K, Jagtap S, Srinivasan SP, Wagh V, Hescheler J, Hengstler J, Leist M, Sachinidis A. Neuronal developmental gene and miRNA signatures induced by histone deacetylase inhibitors in human embryonic stem cells. Cell Death Dis 2015; 6:e1756. [PMID: 25950486 PMCID: PMC4669700 DOI: 10.1038/cddis.2015.121] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 03/26/2015] [Accepted: 03/30/2015] [Indexed: 12/19/2022]
Abstract
Human embryonic stem cells (hESCs) may be applied to develop human-relevant sensitive in vitro test systems for monitoring developmental toxicants. The aim of this study was to identify potential developmental toxicity mechanisms of the histone deacetylase inhibitors (HDAC) valproic acid (VPA), suberoylanilide hydroxamic acid (SAHA) and trichostatin A (TSA) relevant to the in vivo condition using a hESC model in combination with specific differentiation protocols and genome-wide gene expression and microRNA profiling. Analysis of the gene expression data showed that VPA repressed neural tube and dorsal forebrain (OTX2, ISL1, EMX2 and SOX10)-related transcripts. In addition, VPA upregulates axonogenesis and ventral forebrain-associated genes, such as SLIT1, SEMA3A, DLX2/4 and GAD2. HDACi-induced expression of miR-378 and knockdown of miR-378 increases the expression of OTX2 and EMX2, which supports our hypothesis that HDACi targets forebrain markers through miR-378. In conclusion, multilineage differentiation in vitro test system is very sensitive for monitoring molecular activities relevant to in vivo neuronal developmental toxicity. Moreover, miR-378 seems to repress the expression of the OTX2 and EMX2 and therefore could be a regulator of the development of neural tube and dorsal forebrain neurons.
Collapse
Affiliation(s)
- K Meganathan
- Center of Physiology and Pathophysiology, Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), Robert-Koch-Str. 39, Cologne, Germany
| | - S Jagtap
- Center of Physiology and Pathophysiology, Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), Robert-Koch-Str. 39, Cologne, Germany
| | - S P Srinivasan
- Center of Physiology and Pathophysiology, Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), Robert-Koch-Str. 39, Cologne, Germany
| | - V Wagh
- Center of Physiology and Pathophysiology, Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), Robert-Koch-Str. 39, Cologne, Germany
| | - J Hescheler
- Center of Physiology and Pathophysiology, Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), Robert-Koch-Str. 39, Cologne, Germany
| | - J Hengstler
- Leibniz Research Centre for Working Environment and Human Factors, Technical University of Dortmund (IfADo), Dortmund, Germany
| | - M Leist
- Doerenkamp-Zbinden Chair for In Vitro Toxicology and Biomedicine, University of Konstanz, Konstanz, Germany
| | - A Sachinidis
- Center of Physiology and Pathophysiology, Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), Robert-Koch-Str. 39, Cologne, Germany
| |
Collapse
|
41
|
Extra-embryonic Wnt3 regulates the establishment of the primitive streak in mice. Dev Biol 2015; 403:80-8. [PMID: 25907228 DOI: 10.1016/j.ydbio.2015.04.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Revised: 04/10/2015] [Accepted: 04/11/2015] [Indexed: 11/24/2022]
Abstract
The establishment of the head to tail axis at early stages of development is a fundamental aspect of vertebrate embryogenesis. In mice, experimental embryology, genetics and expression studies have suggested that the visceral endoderm, an extra-embryonic tissue, plays an important role in anteroposterior axial development. Here we show that absence of Wnt3 in the posterior visceral endoderm leads to delayed formation of the primitive streak and that interplay between anterior and posterior visceral endoderm restricts the position of the primitive streak. Embryos lacking Wnt3 in the visceral endoderm, however, appear normal by E9.5. Our results suggest a model for axial development in which multiple signals are required for anteroposterior axial development in mammals.
Collapse
|
42
|
Kimura-Yoshida C, Mochida K, Ellwanger K, Niehrs C, Matsuo I. Fate Specification of Neural Plate Border by Canonical Wnt Signaling and Grhl3 is Crucial for Neural Tube Closure. EBioMedicine 2015; 2:513-27. [PMID: 26288816 PMCID: PMC4535158 DOI: 10.1016/j.ebiom.2015.04.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 04/17/2015] [Accepted: 04/17/2015] [Indexed: 12/05/2022] Open
Abstract
During primary neurulation, the separation of a single-layered ectodermal sheet into the surface ectoderm (SE) and neural tube specifies SE and neural ectoderm (NE) cell fates. The mechanisms underlying fate specification in conjunction with neural tube closure are poorly understood. Here, by comparing expression profiles between SE and NE lineages, we observed that uncommitted progenitor cells, expressing stem cell markers, are present in the neural plate border/neural fold prior to neural tube closure. Our results also demonstrated that canonical Wnt and its antagonists, DKK1/KREMEN1, progressively specify these progenitors into SE or NE fates in accord with the progress of neural tube closure. Additionally, SE specification of the neural plate border via canonical Wnt signaling is directed by the grainyhead-like 3 (Grhl3) transcription factor. Thus, we propose that the fate specification of uncommitted progenitors in the neural plate border by canonical Wnt signaling and its downstream effector Grhl3 is crucial for neural tube closure. This study implicates that failure in critical genetic factors controlling fate specification of progenitor cells in the neural plate border/neural fold coordinated with neural tube closure may be potential causes of human neural tube defects. Neural plate border/neural fold possesses stem cell-like characters during primary neurulation. Canonical Wnt and its antagonists progressively specify progenitors into surface or neural fates upon neural tube closure. Fate specification into surface ectoderm in the neural fold is directed by the Grhl3 transcription factor. Fate specification of uncommitted progenitors in the neural plate border is intimately coupled to neural tube closure.
Collapse
Affiliation(s)
- Chiharu Kimura-Yoshida
- Department of Molecular Embryology, Osaka Medical Center, Research Institute for Maternal and Child Health, Osaka Prefectural Hospital Organization, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan
| | - Kyoko Mochida
- Department of Molecular Embryology, Osaka Medical Center, Research Institute for Maternal and Child Health, Osaka Prefectural Hospital Organization, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan
| | - Kristina Ellwanger
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
| | - Christof Niehrs
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany ; Institute of Molecular Biology, 55128 Mainz, Germany
| | - Isao Matsuo
- Department of Molecular Embryology, Osaka Medical Center, Research Institute for Maternal and Child Health, Osaka Prefectural Hospital Organization, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan
| |
Collapse
|
43
|
Ip CK, Fossat N, Jones V, Lamonerie T, Tam PPL. Head formation: OTX2 regulates Dkk1 and Lhx1 activity in the anterior mesendoderm. Development 2014; 141:3859-67. [DOI: 10.1242/dev.114900] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The Otx2 gene encodes a paired-type homeobox transcription factor that is essential for the induction and the patterning of the anterior structures in the mouse embryo. Otx2 knockout embryos fail to form a head. Whereas previous studies have shown that Otx2 is required in the anterior visceral endoderm and the anterior neuroectoderm for head formation, its role in the anterior mesendoderm (AME) has not been assessed specifically. Here, we show that tissue-specific ablation of Otx2 in the AME phenocopies the truncation of the embryonic head of the Otx2 null mutant. Expression of Dkk1 and Lhx1, two genes that are also essential for head formation, is disrupted in the AME of the conditional Otx2-deficient embryos. Consistent with the fact that Dkk1 is a direct target of OTX2, we showed that OTX2 can interact with the H1 regulatory region of Dkk1 to activate its expression. Cross-species comparative analysis, RT-qPCR, ChIP-qPCR and luciferase assays have revealed two conserved regions in the Lhx1 locus to which OTX2 can bind to activate Lhx1 expression. Abnormal development of the embryonic head in Otx2;Lhx1 and Otx2;Dkk1 compound mutant embryos highlights the functional intersection of Otx2, Dkk1 and Lhx1 in the AME for head formation.
Collapse
Affiliation(s)
- Chi Kin Ip
- Embryology Unit, Children's Medical Research Institute, Locked Bag 23, Wentworthville, New South Wales 2145, Australia
- Discipline of Medicine, Sydney Medical School, University of Sydney, New South Wales 2006, Australia
| | - Nicolas Fossat
- Embryology Unit, Children's Medical Research Institute, Locked Bag 23, Wentworthville, New South Wales 2145, Australia
- Discipline of Medicine, Sydney Medical School, University of Sydney, New South Wales 2006, Australia
| | - Vanessa Jones
- Embryology Unit, Children's Medical Research Institute, Locked Bag 23, Wentworthville, New South Wales 2145, Australia
| | - Thomas Lamonerie
- Equipe Neurodéveloppement, Institut de Biologie Valrose, UMR UNS/CNRS 7277/INSERM 1091, Université Nice Sophia Antipolis, Parc Valrose, 06108 Nice cedex 2, France
| | - Patrick P. L. Tam
- Embryology Unit, Children's Medical Research Institute, Locked Bag 23, Wentworthville, New South Wales 2145, Australia
- Discipline of Medicine, Sydney Medical School, University of Sydney, New South Wales 2006, Australia
| |
Collapse
|
44
|
Interleukin-1β-induced Wnt5a enhances human corneal endothelial cell migration through regulation of Cdc42 and RhoA. Mol Cell Biol 2014; 34:3535-45. [PMID: 25022753 DOI: 10.1128/mcb.01572-13] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Wnt5a can activate β-catenin-independent pathways for regulation of various cellular functions, such as migration, that play critical roles in wound repair. Investigation of Wnt5a signaling may help identify therapeutic targets for enhancing corneal endothelial wound healing that could provide an alternative to corneal transplantation in patients with blindness from endothelial dysfunction. However, Wnt5a signaling in corneal endothelial cells (CECs) has not been well characterized. In this study, we show transient induction of Wnt5a by interleukin-1β (IL-1β) stimulation proceeds through NF-κB in human CECs. This leads to binding of Fzd5 to Ror2, resulting in activation of disheveled protein (Dvl) and subsequently disheveled-associated activator of morphogenesis 1 (DAAM1). This leads to activation of Cdc42 and subsequent inhibition of RhoA. Inhibition of RhoA leads to parallel dephosphorylation and inactivation of LIM domain kinase 2 along with dephosphorylation and activation of slingshot 1, resulting in dephosphorylation and activation of cofilin and leading to enhanced cell migration. These findings suggest that Wnt5a enhances cell migration through activation of Cdc42 and inactivation of RhoA in human CECs.
Collapse
|
45
|
Rivera-Pérez JA, Hadjantonakis AK. The Dynamics of Morphogenesis in the Early Mouse Embryo. Cold Spring Harb Perspect Biol 2014; 7:cshperspect.a015867. [PMID: 24968703 DOI: 10.1101/cshperspect.a015867] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
SUMMARYOver the past two decades, our understanding of mouse development from implantation to gastrulation has grown exponentially with an upsurge of genetic, molecular, cellular, and morphogenetic information. New discoveries have exalted the role of extraembryonic tissues in orchestrating embryonic patterning and axial specification. At the same time, the identification of unexpected morphogenetic processes occurring during mouse gastrulation has challenged established dogmas and brought new insights into the mechanisms driving germ layer formation. In this article, we summarize the key findings that have reinvigorated the contemporary view of early postimplantation mammalian development.
Collapse
Affiliation(s)
- Jaime A Rivera-Pérez
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01655
| | - Anna-Katerina Hadjantonakis
- Developmental Biology Program, Sloan-Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| |
Collapse
|
46
|
Ketamine exposure in early development impairs specification of the primary germ cell layers. Neurotoxicol Teratol 2014; 43:59-68. [PMID: 24746641 DOI: 10.1016/j.ntt.2014.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Revised: 04/03/2014] [Accepted: 04/09/2014] [Indexed: 01/25/2023]
Abstract
Preclinical and clinical evidence implicates N-methyl-d-aspartate receptor (NMDAr) signaling in early embryological development. However, the role of NMDAr signaling in early development has not been well studied. Here, we use a mouse embryonic stem cell model to perform a step-wise exploration of the effects of NMDAr signaling on early cell fate specification. We found that antagonism of the NMDAr impaired specification into the neuroectodermal and mesoendodermal cell lineages, with little or no effect on specification of the extraembryonic endoderm cell lineage. Consistent with these findings, exogenous NMDA promoted neuroectodermal differentiation. Finally, NMDAr antagonism modified expression of several key targets of TGF-β superfamily signaling, suggesting a mechanism for these findings. In summary, this study shows that NMDAr antagonism interferes with the normal developmental pathways of embryogenesis, and suggests that interference is most pronounced prior to neuroectodermal and mesoendodermal cell fate specification.
Collapse
|
47
|
Kurokawa D, Ohmura T, Sakurai Y, Inoue K, Suda Y, Aizawa S. Otx2 expression in anterior neuroectoderm and forebrain/midbrain is directed by more than six enhancers. Dev Biol 2014; 387:203-13. [PMID: 24457099 DOI: 10.1016/j.ydbio.2014.01.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Revised: 01/12/2014] [Accepted: 01/14/2014] [Indexed: 11/25/2022]
Abstract
Otx2 plays essential roles in each site at each step of head development. We previously identified the AN1 enhancer at 91kb 5' upstream for the Otx2 expressions in anterior neuroectoderm (AN) at neural plate stage before E8.5, and the FM1 enhancer at 75kb 5' upstream and the FM2 enhancer at 122kb 3' downstream for the expression in forebrain/midbrain (FM) at brain vesicle stage after E8.5. The present study identified a second AN enhancer (AN2) at 88kb 5' upstream; the AN2 enhancer also recapitulates the endogenous Otx2 expression in choroid plexus, cortical hem and choroidal roof. However, the enhancer mutants indicated the presence of another AN enhancer. The study also identified a third FM enhancer (FM3) at 153kb 5' upstream. Thus, the Otx2 expressions in anterior neuroectoderm and forebrain/midbrain are regulated by more than six enhancers located far from the coding region. The enhancers identified are differentially conserved among vertebrates; none of the AN enhancers has activities in caudal forebrain and midbrain at brain vesicle stage after E8.5, nor do any of the FM enhancers in anterior neuroectoderm at neural plate stage before E8.5.
Collapse
Affiliation(s)
- Daisuke Kurokawa
- Laboratory for Vertebrate Body Plan, Center for Developmental Biology (CDB), RIKEN Kobe, 2-2-3 Minatojima Minami-machi, Chuo-Ku, Kobe, Hyogo 650-0047, Japan; Misaki Marine Biological Station, Graduate School of Science, The University of Tokyo, 1024 Koajiro, Misaki, Miura, Kanagawa 238-0225, Japan
| | - Tomomi Ohmura
- Laboratory for Vertebrate Body Plan, Center for Developmental Biology (CDB), RIKEN Kobe, 2-2-3 Minatojima Minami-machi, Chuo-Ku, Kobe, Hyogo 650-0047, Japan
| | - Yusuke Sakurai
- Laboratory for Vertebrate Body Plan, Center for Developmental Biology (CDB), RIKEN Kobe, 2-2-3 Minatojima Minami-machi, Chuo-Ku, Kobe, Hyogo 650-0047, Japan
| | - Kenichi Inoue
- Laboratory for Animal Resources and Genetic Engineering, Center for Developmental Biology (CDB), RIKEN Kobe, 2-2-3 Minatojima Minami-machi, Chuo-Ku, Kobe, Hyogo 650-0047, Japan
| | - Yoko Suda
- Laboratory for Vertebrate Body Plan, Center for Developmental Biology (CDB), RIKEN Kobe, 2-2-3 Minatojima Minami-machi, Chuo-Ku, Kobe, Hyogo 650-0047, Japan
| | - Shinichi Aizawa
- Laboratory for Vertebrate Body Plan, Center for Developmental Biology (CDB), RIKEN Kobe, 2-2-3 Minatojima Minami-machi, Chuo-Ku, Kobe, Hyogo 650-0047, Japan; Laboratory for Animal Resources and Genetic Engineering, Center for Developmental Biology (CDB), RIKEN Kobe, 2-2-3 Minatojima Minami-machi, Chuo-Ku, Kobe, Hyogo 650-0047, Japan.
| |
Collapse
|
48
|
Hiramatsu R, Matsuoka T, Kimura-Yoshida C, Han SW, Mochida K, Adachi T, Takayama S, Matsuo I. External mechanical cues trigger the establishment of the anterior-posterior axis in early mouse embryos. Dev Cell 2014; 27:131-144. [PMID: 24176640 DOI: 10.1016/j.devcel.2013.09.026] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 07/16/2013] [Accepted: 09/27/2013] [Indexed: 10/26/2022]
Abstract
Mouse anterior-posterior axis polarization is preceded by formation of the distal visceral endoderm (DVE) by unknown mechanisms. Here, we show by in vitro culturing of embryos immediately after implantation in microfabricated cavities that the external mechanical cues exerted on the embryo are crucial for DVE formation, as well as the elongated egg cylinder shape, without affecting embryo-intrinsic transcriptional programs except those involving DVE-specific genes. This implies that these developmental events immediately after implantation are not simply embryo-autonomous processes but require extrinsic factors from maternal tissues. Moreover, the mechanical forces induce a breach of the basement membrane barrier at the distal portion locally, and thereby the transmigrated epiblast cells emerge as the DVE cells. Thus, we propose that external mechanical forces exerted by the interaction between embryo and maternal uterine tissues directly control the location of DVE formation at the distal tip and consequently establish the mammalian primary body axis.
Collapse
Affiliation(s)
- Ryuji Hiramatsu
- Department of Molecular Embryology, Osaka Medical Center and Research Institute for Maternal and Child Health, Osaka Prefectural Hospital Organization, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan; Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, 4-7-1 Gakuen, Musashi-Murayama, Tokyo 208-0011, Japan
| | - Toshiki Matsuoka
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Chiharu Kimura-Yoshida
- Department of Molecular Embryology, Osaka Medical Center and Research Institute for Maternal and Child Health, Osaka Prefectural Hospital Organization, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan
| | - Sung-Woong Han
- Department of Biomechanics, Institute for Frontier Medical Sciences, Kyoto University, 53 Kawaharacho, Shogoin, Sakyo, Kyoto 606-8507, Japan
| | - Kyoko Mochida
- Department of Molecular Embryology, Osaka Medical Center and Research Institute for Maternal and Child Health, Osaka Prefectural Hospital Organization, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan
| | - Taiji Adachi
- Department of Biomechanics, Institute for Frontier Medical Sciences, Kyoto University, 53 Kawaharacho, Shogoin, Sakyo, Kyoto 606-8507, Japan
| | - Shuichi Takayama
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Isao Matsuo
- Department of Molecular Embryology, Osaka Medical Center and Research Institute for Maternal and Child Health, Osaka Prefectural Hospital Organization, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan; Department of Pediatric and Neonatal-Perinatal Research, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
49
|
Trott J, Martinez Arias A. Single cell lineage analysis of mouse embryonic stem cells at the exit from pluripotency. Biol Open 2013; 2:1049-56. [PMID: 24167715 PMCID: PMC3798188 DOI: 10.1242/bio.20135934] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2013] [Accepted: 07/15/2013] [Indexed: 12/29/2022] Open
Abstract
Understanding how interactions between extracellular signalling pathways and transcription factor networks influence cellular decision making will be crucial for understanding mammalian embryogenesis and for generating specialised cell types in vitro. To this end, pluripotent mouse Embryonic Stem (mES) cells have proven to be a useful model system. However, understanding how transcription factors and signalling pathways affect decisions made by individual cells is confounded by the fact that measurements are generally made on groups of cells, whilst individual mES cells differentiate at different rates and towards different lineages, even in conditions that favour a particular lineage. Here we have used single-cell measurements of transcription factor expression and Wnt/β-catenin signalling activity to investigate their effects on lineage commitment decisions made by individual cells. We find that pluripotent mES cells exhibit differing degrees of heterogeneity in their expression of important regulators from pluripotency, depending on the signalling environment to which they are exposed. As mES cells differentiate, downregulation of Nanog and Oct4 primes cells for neural commitment, whilst loss of Sox2 expression primes cells for primitive streak commitment. Furthermore, we find that Wnt signalling acts through Nanog to direct cells towards a primitive streak fate, but that transcriptionally active β-catenin is associated with both neural and primitive streak commitment. These observations confirm and extend previous suggestions that pluripotency genes influence lineage commitment and demonstrate how their dynamic expression affects the direction of lineage commitment, whilst illustrating two ways in which the Wnt signalling pathway acts on this network during cell fate assignment.
Collapse
Affiliation(s)
- Jamie Trott
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
- Wellcome Trust Centre for Stem Cell Research, University of Cambridge, Cambridge CB2 1QR, UK
- Present address: Institute of Medical Biology, 8A Biomedical Grove, No. 06-06 Immunos, Singapore 138648
| | | |
Collapse
|
50
|
Ets2-dependent trophoblast signalling is required for gastrulation progression after primitive streak initiation. Nat Commun 2013; 4:1658. [DOI: 10.1038/ncomms2646] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 02/25/2013] [Indexed: 11/09/2022] Open
|