1
|
Ren H, Jia X, Yu L. The building blocks of embryo models: embryonic and extraembryonic stem cells. Cell Discov 2025; 11:40. [PMID: 40258839 PMCID: PMC12012135 DOI: 10.1038/s41421-025-00780-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 01/10/2025] [Indexed: 04/23/2025] Open
Abstract
The process of a single-celled zygote developing into a complex multicellular organism is precisely regulated at spatial and temporal levels in vivo. However, understanding the mechanisms underlying development, particularly in humans, has been constrained by technical and ethical limitations associated with studying natural embryos. Harnessing the intrinsic ability of embryonic stem cells (ESCs) to self-organize when induced and assembled, researchers have established several embryo models as alternative approaches to studying early development in vitro. Recent studies have revealed the critical role of extraembryonic cells in early development; and many groups have created more sophisticated and precise ESC-derived embryo models by incorporating extraembryonic stem cell lines, such as trophoblast stem cells (TSCs), extraembryonic mesoderm cells (EXMCs), extraembryonic endoderm cells (XENs, in rodents), and hypoblast stem cells (in primates). Here, we summarize the characteristics of existing mouse and human embryonic and extraembryonic stem cells and review recent advancements in developing mouse and human embryo models.
Collapse
Affiliation(s)
- Hongan Ren
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaojie Jia
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Leqian Yu
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
2
|
Nakashima Y, Tsukahara M. MFGE8 Acts as a Cell Adhesion Factor for Human-Induced Pluripotent Stem Cells in Embryology. Tissue Eng Part C Methods 2025; 31:74-84. [PMID: 39869126 DOI: 10.1089/ten.tec.2024.0328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025] Open
Abstract
Mouse embryonic fibroblasts (MEFs) have been widely used as feeder cells in embryonic stem cell cultures because they can mimic the embryonic microenvironment. Milk fat globule-epidermal growth factor 8 (MFGE8) is expressed during mouse gonadal development, 10.5-13.5 embryonic, and is also found in MEF-conditioned medium (MEF-CM). Feeder-less culture of human-induced pluripotent stem cells (iPSCs) with MEF-CM significantly decreased the number of adherent cells when an inhibitory antibody against MFGE8 was used. The concentration of mouse MFGE8 in MEF-CM, as measured by an ELISA (Enzyme-Linked Immunosorbent Assay), was 0.16-1.24 μg/mL. Mouse MFGE8 and human MFGE8 have partially different molecular structures. Both the recombinant mouse MFGE8 and human MFGE8 significantly promoted cell adhesion of human iPSCs at medium-added concentrations of 2 μg/mL. This cell adhesion was also strongly inhibited by Arginylglycylaspartic acid (RGD) inhibitors, suggesting that it is dependent on the RGD sequence. The integrin αVβ5 expressed in iPSCs was thought to be involved in binding to the RGD sequence. MEF-CMs have long been an essential bio-derived material for the feeder culture method of iPSC culture. This study demonstrates that MFGE8 in MEF-CM is a functional factor in the promoting of cell adhesion of human iPSCs. Furthermore, the use of MFGE8-containing media demonstrates that iPSCs can be established and cultured while maintaining pluripotency and inducing three germ layer differentiation. The results of this study suggest the possibility of using MFGE8 as a scaffold material suitable for inducing differentiation when reproducing in vivo maturation in vitro.
Collapse
Affiliation(s)
- Yoshiki Nakashima
- CiRA Foundation, Research and Development Center, Osaka, Japan
- Research and Development Center, Kyoto University Center for iPS Cell Research and Application Foundation (CiRA Foundation), Kyoto, Japan
| | | |
Collapse
|
3
|
Ying Q, Nichols J. Relationship of PSC to embryos: Extending and refining capture of PSC lines from mammalian embryos. Bioessays 2024; 46:e2400077. [PMID: 39400400 PMCID: PMC11589693 DOI: 10.1002/bies.202400077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 09/07/2024] [Indexed: 10/15/2024]
Abstract
Pluripotent stem cell lines derived from preimplantation mouse embryos have opened opportunities for the study of early mammalian development and generation of genetically uncompromised material for differentiation into specific cell types. Murine embryonic stem cells are highly versatile and can be engineered and introduced into host embryos, transferred to recipient females, and gestated to investigate gene function at multiple levels as well as developmental mechanisms, including lineage segregation and cell competition. In this review, we summarize the biomedical motivation driving the incremental modification to culture regimes and analyses that have advanced stem cell research to its current state. Ongoing investigation into divergent mechanisms of early developmental processes adopted by other species, such as agriculturally beneficial mammals and birds, will continue to enrich knowledge and inform strategies for future in vitro models.
Collapse
Affiliation(s)
- Qi‐Long Ying
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Jennifer Nichols
- MRC Human Genetics Unit, Institute for Genetics and CancerUniversity of EdinburghEdinburghUK
| |
Collapse
|
4
|
Smith A. Propagating pluripotency - The conundrum of self-renewal. Bioessays 2024; 46:e2400108. [PMID: 39180242 PMCID: PMC11589686 DOI: 10.1002/bies.202400108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/29/2024] [Accepted: 08/06/2024] [Indexed: 08/26/2024]
Abstract
The discovery of mouse embryonic stem cells in 1981 transformed research in mammalian developmental biology and functional genomics. The subsequent generation of human pluripotent stem cells (PSCs) and the development of molecular reprogramming have opened unheralded avenues for drug discovery and cell replacement therapy. Here, I review the history of PSCs from the perspective that long-term self-renewal is a product of the in vitro signaling environment, rather than an intrinsic feature of embryos. I discuss the relationship between pluripotent states captured in vitro to stages of epiblast in the embryo and suggest key considerations for evaluation of PSCs. A remaining fundamental challenge is to determine whether naïve pluripotency can be propagated from the broad range of mammals by exploiting common principles in gene regulatory architecture.
Collapse
Affiliation(s)
- Austin Smith
- Living Systems InstituteUniversity of ExeterExeterUK
| |
Collapse
|
5
|
Chen R, Fan R, Chen F, Govindasamy N, Brinkmann H, Stehling M, Adams RH, Jeong HW, Bedzhov I. Analyzing embryo dormancy at single-cell resolution reveals dynamic transcriptional responses and activation of integrin-Yap/Taz prosurvival signaling. Cell Stem Cell 2024; 31:1262-1279.e8. [PMID: 39047740 PMCID: PMC7617458 DOI: 10.1016/j.stem.2024.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 05/03/2024] [Accepted: 06/28/2024] [Indexed: 07/27/2024]
Abstract
Embryonic diapause is a reproductive adaptation that enables some mammalian species to halt the otherwise continuous pace of embryonic development. In this dormant state, the embryo exploits poorly understood regulatory mechanisms to preserve its developmental potential for prolonged periods of time. Here, using mouse embryos and single-cell RNA sequencing, we molecularly defined embryonic diapause at single-cell resolution, revealing transcriptional dynamics while the embryo seemingly resides in a state of suspended animation. Additionally, we found that the dormant pluripotent cells rely on integrin receptors to sense their microenvironment and preserve their viability via Yap/Taz-mediated prosurvival signaling.
Collapse
Affiliation(s)
- Rui Chen
- Embryonic Self-Organization Research Group, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Rui Fan
- Embryonic Self-Organization Research Group, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Fei Chen
- Embryonic Self-Organization Research Group, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Niraimathi Govindasamy
- Embryonic Self-Organization Research Group, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Heike Brinkmann
- Embryonic Self-Organization Research Group, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Martin Stehling
- Flow Cytometry Unit, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany
| | - Hyun-Woo Jeong
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany; Single Cell Multi-Omics Laboratory, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany.
| | - Ivan Bedzhov
- Embryonic Self-Organization Research Group, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, 48149 Münster, Germany.
| |
Collapse
|
6
|
Zhang J, Tong L, Liu Y, Li X, Wang J, Lin R, Zhou Z, Chen Y, Chen Y, Liu Y, Chen D. The regulatory role of m 6A modification in the maintenance and differentiation of embryonic stem cells. Genes Dis 2024; 11:101199. [PMID: 38947741 PMCID: PMC11214295 DOI: 10.1016/j.gendis.2023.101199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/28/2023] [Accepted: 11/11/2023] [Indexed: 07/02/2024] Open
Abstract
As the most prevalent and reversible internal epigenetic modification in eukaryotic mRNAs, N 6-methyladenosine (m6A) post-transcriptionally regulates the processing and metabolism of mRNAs involved in diverse biological processes. m6A modification is regulated by m6A writers, erasers, and readers. Emerging evidence suggests that m6A modification plays essential roles in modulating the cell-fate transition of embryonic stem cells. Mechanistic investigation of embryonic stem cell maintenance and differentiation is critical for understanding early embryonic development, which is also the premise for the application of embryonic stem cells in regenerative medicine. This review highlights the current knowledge of m6A modification and its essential regulatory contribution to the cell fate transition of mouse and human embryonic stem cells.
Collapse
Affiliation(s)
- Jin Zhang
- Center for Reproductive Medicine of the Second Affiliated Hospital, Center for Regeneration and Cell Therapy of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Lingling Tong
- Center for Reproductive Medicine of the Second Affiliated Hospital, Center for Regeneration and Cell Therapy of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Yuchen Liu
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Xiang Li
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Jiayi Wang
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Ruoxin Lin
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Ziyu Zhou
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Yunbing Chen
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Yanxi Chen
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Yirong Liu
- College of Materials and Chemical Engineering, Minjiang University, Fuzhou, Fujian 350108, China
| | - Di Chen
- Center for Reproductive Medicine of the Second Affiliated Hospital, Center for Regeneration and Cell Therapy of Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
- National Key Laboratory of Biobased Transportation Fuel Technology, Haining, Zhejiang 314400, China
| |
Collapse
|
7
|
Ghorbani S, Christine Füchtbauer A, Møllebjerg A, Møller Martensen P, Hvidbjerg Laursen S, Christian Evar Kraft D, Kjems J, Meyer RL, Rahimi K, Foss M, Füchtbauer EM, Sutherland DS. Protein ligand and nanotopography separately drive the phenotype of mouse embryonic stem cells. Biomaterials 2023; 301:122244. [PMID: 37459700 DOI: 10.1016/j.biomaterials.2023.122244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 07/09/2023] [Accepted: 07/12/2023] [Indexed: 09/06/2023]
Abstract
Biochemical and biomechanical signals regulate stem cell function in the niche environments in vivo. Current in vitro culture of mouse embryonic stem cells (mESC) uses laminin (LN-511) to provide mimetic biochemical signaling (LN-521 for human systems) to maintain stemness. Alternative approaches propose topographical cues to provide biomechanical cues, however combined biochemical and topographic cues may better mimic the in vivo environment, but are largely unexplored for in vitro stem cell expansion. In this study, we directly compare in vitro signals from LN-511 and/or topographic cues to maintain stemness, using systematically-varied submicron pillar patterns or flat surfaces with or without preadsorbed LN-511. The adhesion of cells, colony formation, expression of the pluripotency marker,octamer-binding transcription factor 4 (Oct4), and transcriptome profiling were characterized. We observed that either biochemical or topographic signals could maintain stemness of mESCs in feeder-free conditions, indicated by high-level Oct4 and gene profiling by RNAseq. The combination of LN-511 with nanotopography reduced colony growth, while maintaining stemness markers, shifted the cellular phenotype indicating that the integration of biochemical and topographic signals is antagonistic. Overall, significantly faster (up to 2.5 times) colony growth was observed at nanotopographies without LN-511, suggesting for improved ESC expansion.
Collapse
Affiliation(s)
- Sadegh Ghorbani
- Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark; The Centre for Cellular Signal Patterns (CELLPAT), Gustav Wieds Vej 14, Aarhus C, 8000, Denmark; Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA.
| | | | - Andreas Møllebjerg
- Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark
| | | | - Sara Hvidbjerg Laursen
- Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark
| | - David Christian Evar Kraft
- Department of Dentistry and Oral Health, Faculty of Health, University of Aarhus, Aarhus C, 8000, Denmark
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark; The Centre for Cellular Signal Patterns (CELLPAT), Gustav Wieds Vej 14, Aarhus C, 8000, Denmark; Department of Molecular Biology, University of Aarhus, Aarhus C, 8000, Denmark
| | - Rikke Louise Meyer
- Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark
| | - Karim Rahimi
- Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark; Department of Molecular Biology, University of Aarhus, Aarhus C, 8000, Denmark
| | - Morten Foss
- Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark
| | | | - Duncan S Sutherland
- Interdisciplinary Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, Aarhus C, 8000, Denmark; The Centre for Cellular Signal Patterns (CELLPAT), Gustav Wieds Vej 14, Aarhus C, 8000, Denmark.
| |
Collapse
|
8
|
New insights into the epitranscriptomic control of pluripotent stem cell fate. Exp Mol Med 2022; 54:1643-1651. [PMID: 36266446 PMCID: PMC9636187 DOI: 10.1038/s12276-022-00824-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/26/2022] [Accepted: 06/01/2022] [Indexed: 12/29/2022] Open
Abstract
Each cell in the human body has a distinguishable fate. Pluripotent stem cells are challenged with a myriad of lineage differentiation options. Defects are more likely to be fatal to stem cells than to somatic cells due to the broad impact of the former on early development. Hence, a detailed understanding of the mechanisms that determine the fate of stem cells is needed. The mechanisms by which human pluripotent stem cells, although not fully equipped with complex chromatin structures or epigenetic regulatory mechanisms, accurately control gene expression and are important to the stem cell field. In this review, we examine the events driving pluripotent stem cell fate and the underlying changes in gene expression during early development. In addition, we highlight the role played by the epitranscriptome in the regulation of gene expression that is necessary for each fate-related event.
Collapse
|
9
|
Kim J, Muraoka M, Okada H, Toyoda A, Ajima R, Saga Y. The RNA helicase DDX6 controls early mouse embryogenesis by repressing aberrant inhibition of BMP signaling through miRNA-mediated gene silencing. PLoS Genet 2022; 18:e1009967. [PMID: 36197846 PMCID: PMC9534413 DOI: 10.1371/journal.pgen.1009967] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 08/11/2022] [Indexed: 11/29/2022] Open
Abstract
The evolutionarily conserved RNA helicase DDX6 is a central player in post-transcriptional regulation, but its role during embryogenesis remains elusive. We here show that DDX6 enables proper cell lineage specification from pluripotent cells by analyzing Ddx6 knockout (KO) mouse embryos and employing an in vitro epiblast-like cell (EpiLC) induction system. Our study unveils that DDX6 is an important BMP signaling regulator. Deletion of Ddx6 causes the aberrant upregulation of the negative regulators of BMP signaling, which is accompanied by enhanced expression of Nodal and related genes. Ddx6 KO pluripotent cells acquire higher pluripotency with a strong inclination toward neural lineage commitment. During gastrulation, abnormally expanded Nodal and Eomes expression in the primitive streak likely promotes endoderm cell fate specification while inhibiting mesoderm differentiation. We also genetically dissected major DDX6 pathways by generating Dgcr8, Dcp2, and Eif4enif1 KO models in addition to Ddx6 KO. We found that the miRNA pathway mutant Dgcr8 KO phenocopies Ddx6 KO, indicating that DDX6 mostly works along with the miRNA pathway during early development, whereas its P-body-related functions are dispensable. Therefore, we conclude that DDX6 prevents aberrant upregulation of BMP signaling inhibitors by participating in miRNA-mediated gene silencing processes. Overall, this study delineates how DDX6 affects the development of the three primary germ layers during early mouse embryogenesis and the underlying mechanism of DDX6 function.
Collapse
Affiliation(s)
- Jessica Kim
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Masafumi Muraoka
- Mammalian Development Laboratory, Department of Gene Function and Phenomics, National Institute of Genetics, Mishima, Japan
| | - Hajime Okada
- Mammalian Development Laboratory, Department of Gene Function and Phenomics, National Institute of Genetics, Mishima, Japan
| | - Atsushi Toyoda
- Advanced Genomics Center, National Institute of Genetics, Mishima, Japan
| | - Rieko Ajima
- Mammalian Development Laboratory, Department of Gene Function and Phenomics, National Institute of Genetics, Mishima, Japan
- Department of Genetics, The Graduate University for Advanced Studies, SOKENDAI, Mishima, Japan
- * E-mail: (RA); (YS)
| | - Yumiko Saga
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Tokyo, Japan
- Mammalian Development Laboratory, Department of Gene Function and Phenomics, National Institute of Genetics, Mishima, Japan
- Department of Genetics, The Graduate University for Advanced Studies, SOKENDAI, Mishima, Japan
- * E-mail: (RA); (YS)
| |
Collapse
|
10
|
Wang J, Xiao B, Kimura E, Mongan M, Xia Y. The combined effects of Map3k1 mutation and dioxin on differentiation of keratinocytes derived from mouse embryonic stem cells. Sci Rep 2022; 12:11482. [PMID: 35798792 PMCID: PMC9263165 DOI: 10.1038/s41598-022-15760-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 06/29/2022] [Indexed: 11/25/2022] Open
Abstract
Epithelial development starts with stem cell commitment to ectoderm followed by differentiation to the basal keratinocytes. The basal keratinocytes, first committed in embryogenesis, constitute the basal layer of the epidermis. They have robust proliferation and differentiation potential and are responsible for epidermal expansion, maintenance and regeneration. We generated basal epithelial cells in vitro through differentiation of mouse embryonic stem cells (mESCs). Early on in differentiation, the expression of stem cell markers, Oct4 and Nanog, decreased sharply along with increased ectoderm marker keratin (Krt) 18. Later on, Krt 18 expression was subdued when cells displayed basal keratinocyte characteristics, including regular polygonal shape, adherent and tight junctions and Krt 14 expression. These cells additionally expressed abundant Sca-1, Krt15 and p63, suggesting epidermal progenitor characteristics. Using Map3k1 mutant mESCs and environmental dioxin, we examined the gene and environment effects on differentiation. Neither Map3k1 mutation nor dioxin altered mESC differentiation to ectoderm and basal keratinocytes, but they, individually and in combination, potentiated Krt 1 expression and basal to spinous differentiation. Similar gene-environment effects were observed in vivo where dioxin exposure increased Krt 1 more substantially in the epithelium of Map3k1+/- than wild type embryos. Thus, the in vitro model of epithelial differentiation can be used to investigate the effects of genetic and environmental factors on epidermal development.
Collapse
Affiliation(s)
- Jingjing Wang
- Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, Cincinnati, OH, 45267-0056, USA
| | - Bo Xiao
- Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, Cincinnati, OH, 45267-0056, USA
| | - Eiki Kimura
- Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, Cincinnati, OH, 45267-0056, USA
| | - Maureen Mongan
- Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, Cincinnati, OH, 45267-0056, USA
| | - Ying Xia
- Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, Cincinnati, OH, 45267-0056, USA.
| |
Collapse
|
11
|
Rostovskaya M, Andrews S, Reik W, Rugg-Gunn PJ. Amniogenesis occurs in two independent waves in primates. Cell Stem Cell 2022; 29:744-759.e6. [PMID: 35439430 DOI: 10.1016/j.stem.2022.03.014] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 02/16/2022] [Accepted: 03/24/2022] [Indexed: 01/28/2023]
Abstract
In primates, the amnion emerges through cavitation of the epiblast during implantation, whereas in other species it does so later at gastrulation by the folding of the ectoderm. How the mechanisms of amniogenesis diversified during evolution remains unknown. Unexpectedly, single-cell analysis of primate embryos uncovered two transcriptionally and temporally distinct amniogenesis waves. To study this, we employed the naive-to-primed transition of human pluripotent stem cells (hPSCs) to model peri-implantation epiblast development. Partially primed hPSCs transiently gained the ability to differentiate into cavitating epithelium that transcriptionally and morphologically matched the early amnion, whereas fully primed hPSCs produced cells resembling the late amnion instead, thus recapitulating the two independent differentiation waves. The early wave follows a trophectoderm-like pathway and encompasses cavitation, whereas the late wave resembles an ectoderm-like route during gastrulation. The discovery of two independent waves explains how amniogenesis through cavitation could emerge during evolution via duplication of the pre-existing trophectoderm program.
Collapse
Affiliation(s)
| | - Simon Andrews
- Bioinformatics Group, Babraham Institute, Cambridge CB22 3AT, UK
| | - Wolf Reik
- Epigenetics Programme, Babraham Institute, Cambridge CB22 3AT, UK; Altoslabs Cambridge Institute, Cambridge CB21 6GP, UK; Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1QR, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK; Wellcome-MRC Stem Cell Institute, Cambridge CB2 0AW, UK.
| | - Peter J Rugg-Gunn
- Epigenetics Programme, Babraham Institute, Cambridge CB22 3AT, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK; Wellcome-MRC Stem Cell Institute, Cambridge CB2 0AW, UK.
| |
Collapse
|
12
|
Alonso-Alonso S, Santaló J, Ibáñez E. Efficient generation of embryonic stem cells from single blastomeres of cryopreserved mouse embryos in the presence of signalling modulators. Reprod Fertil Dev 2022; 34:576-587. [PMID: 35157826 DOI: 10.1071/rd21297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/23/2022] [Indexed: 11/23/2022] Open
Abstract
CONTEXT Derivation of embryonic stem cells (ESC) from single blastomeres is an interesting alternative to the use of whole blastocysts, but derivation rates are lower and the requirements for successful ESC obtention are still poorly defined. AIMS To investigate the effects of embryo cryopreservation and of signalling modulators present during embryo culture and/or ESC establishment on ESC derivation efficiency from single 8-cell mouse blastomeres. METHOD Fresh and cryopreserved 2-cell embryos were cultured and biopsied at the 8-cell stage. Single blastomeres were cultured in the presence of 2i or R2i cocktails, with or without adrenocorticotropic hormone (ACTH). We analysed ESC derivation efficiencies and characterised pluripotency genes expression and karyotype integrity of the resulting lines. We also evaluated the impact of embryo preculture with R2i on epiblast cell numbers and derivation rates. KEY RESULTS The ESC generation was not compromised by embryo cryopreservation and ACTH was dispensable under most of the conditions tested. While 2i and R2i were similarly effective for ESC derivation, R2i provided higher karyotype integrity. Embryo preculture with R2i yielded increased numbers of epiblast cells but did not lead to increased ESC generation. CONCLUSIONS Our findings help to define a simplified and efficient procedure for the establishment of mouse ESC from single 8-cell blastomeres. IMPLICATIONS This study will contribute to improving the potential of this experimental procedure, providing a tool to investigate the developmental potential of blastomeres isolated from different embryonic stages and to reduce the number of embryos needed for ESC derivation.
Collapse
Affiliation(s)
- Sandra Alonso-Alonso
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Josep Santaló
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Elena Ibáñez
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biociències, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| |
Collapse
|
13
|
Pedone E, Failli M, Gambardella G, De Cegli R, La Regina A, di Bernardo D, Marucci L. β-catenin perturbations control differentiation programs in mouse embryonic stem cells. iScience 2022; 25:103756. [PMID: 35128356 PMCID: PMC8804270 DOI: 10.1016/j.isci.2022.103756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 11/09/2021] [Accepted: 01/07/2022] [Indexed: 11/06/2022] Open
Abstract
The Wnt/β-catenin pathway is involved in development, cancer, and embryonic stem cell (ESC) maintenance; its dual role in stem cell self-renewal and differentiation is still controversial. Here, by applying an in vitro system enabling inducible gene expression control, we report that moderate induction of transcriptionally active exogenous β-catenin in β-catenin null mouse ESCs promotes epiblast-like cell (EpiLC) derivation in vitro. Instead, in wild-type cells, moderate chemical pre-activation of the Wnt/β-catenin pathway promotes EpiLC in vitro derivation. Finally, we suggest that moderate β-catenin levels in β-catenin null mouse ESCs favor early stem cell commitment toward mesoderm if the exogenous protein is induced only in the “ground state” of pluripotency condition, or endoderm if the induction is maintained during the differentiation. Overall, our results confirm previous findings about the role of β-catenin in pluripotency and differentiation, while indicating a role for its doses in promoting specific differentiation programs. Moderate β-catenin levels promote EpiLCs derivation in vitro Chemical pre-activation of the Wnt pathway enhances ESC-EpiLC transition β-catenin overexpression tips the balance between mesoderm and endoderm Cell fate is influenced by the extent of β-catenin induction
Collapse
|
14
|
Abstract
Naïve pluripotent stem cells are the in vitro counterparts of pre-implantation embryonic epiblast. During the last few years, several protocols for establishing and maintaining human pluripotent stem cells (hPSCs) with naïve features have been reported, and many of these protocols result in cell populations with different molecular characteristics. As such, choosing the most appropriate method for naïve hPSC maintenance can pose a significant challenge. This chapter presents an optimized system called PXGL for culturing naïve hPSCs. Naïve hPSCs robustly self-renew while retaining a normal karyotype in PXGL, and the protocol is reproducible across different cell lines and independent laboratories.
Collapse
|
15
|
OUP accepted manuscript. Stem Cells 2022; 40:751-762. [DOI: 10.1093/stmcls/sxac034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 04/22/2022] [Indexed: 11/13/2022]
|
16
|
Rostovskaya M. Capacitation of Human Naïve Pluripotent Stem Cells. Methods Mol Biol 2022; 2416:117-131. [PMID: 34870834 DOI: 10.1007/978-1-0716-1908-7_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Naïve and primed pluripotent stem cells resemble epiblast cells of the pre-implantation and post-implantation embryo, respectively. This chapter describes a simple experimental system for the efficient and consistent transition of human pluripotent stem cells (hPSCs) from the naïve to the primed state, which is a process called capacitation. Naïve hPSCs after capacitation can be differentiated further to somatic lineages, thus reproducing the order of developmental events in the embryo. Protocols for the induction of neuroectoderm, definitive endoderm, and paraxial mesoderm from hPSCs after capacitation and also from conventionally derived primed hPSCs are included in the chapter. Importantly, hPSC capacitation closely recapitulates transcriptional, metabolic, signaling, and cell polarity changes in the epiblast of primate embryos, and therefore offers a unique in vitro model of human peri-implantation development.
Collapse
|
17
|
Yeh CY, Huang WH, Chen HC, Meir YJJ. Capturing Pluripotency and Beyond. Cells 2021; 10:cells10123558. [PMID: 34944066 PMCID: PMC8700150 DOI: 10.3390/cells10123558] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 11/16/2022] Open
Abstract
During the development of a multicellular organism, the specification of different cell lineages originates in a small group of pluripotent cells, the epiblasts, formed in the preimplantation embryo. The pluripotent epiblast is protected from premature differentiation until exposure to inductive cues in strictly controlled spatially and temporally organized patterns guiding fetus formation. Epiblasts cultured in vitro are embryonic stem cells (ESCs), which recapitulate the self-renewal and lineage specification properties of their endogenous counterparts. The characteristics of totipotency, although less understood than pluripotency, are becoming clearer. Recent studies have shown that a minor ESC subpopulation exhibits expanded developmental potential beyond pluripotency, displaying a characteristic reminiscent of two-cell embryo blastomeres (2CLCs). In addition, reprogramming both mouse and human ESCs in defined media can produce expanded/extended pluripotent stem cells (EPSCs) similar to but different from 2CLCs. Further, the molecular roadmaps driving the transition of various potency states have been clarified. These recent key findings will allow us to understand eutherian mammalian development by comparing the underlying differences between potency network components during development. Using the mouse as a paradigm and recent progress in human PSCs, we review the epiblast's identity acquisition during embryogenesis and their ESC counterparts regarding their pluripotent fates and beyond.
Collapse
Affiliation(s)
- Chih-Yu Yeh
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (C.-Y.Y.); (W.-H.H.)
| | - Wei-Han Huang
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (C.-Y.Y.); (W.-H.H.)
| | - Hung-Chi Chen
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (C.-Y.Y.); (W.-H.H.)
- Limbal Stem Cell Laboratory, Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou 333, Taiwan
- Correspondence: (H.-C.C.); (Y.-J.J.M.)
| | - Yaa-Jyuhn James Meir
- Limbal Stem Cell Laboratory, Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou 333, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Correspondence: (H.-C.C.); (Y.-J.J.M.)
| |
Collapse
|
18
|
van der Weijden VA, Bulut-Karslioglu A. Molecular Regulation of Paused Pluripotency in Early Mammalian Embryos and Stem Cells. Front Cell Dev Biol 2021; 9:708318. [PMID: 34386497 PMCID: PMC8353277 DOI: 10.3389/fcell.2021.708318] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/06/2021] [Indexed: 02/06/2023] Open
Abstract
The energetically costly mammalian investment in gestation and lactation requires plentiful nutritional sources and thus links the environmental conditions to reproductive success. Flexibility in adjusting developmental timing enhances chances of survival in adverse conditions. Over 130 mammalian species can reversibly pause early embryonic development by switching to a near dormant state that can be sustained for months, a phenomenon called embryonic diapause. Lineage-specific cells are retained during diapause, and they proliferate and differentiate upon activation. Studying diapause thus reveals principles of pluripotency and dormancy and is not only relevant for development, but also for regeneration and cancer. In this review, we focus on the molecular regulation of diapause in early mammalian embryos and relate it to maintenance of potency in stem cells in vitro. Diapause is established and maintained by active rewiring of the embryonic metabolome, epigenome, and gene expression in communication with maternal tissues. Herein, we particularly discuss factors required at distinct stages of diapause to induce, maintain, and terminate dormancy.
Collapse
|
19
|
Guo G, Stirparo GG, Strawbridge SE, Spindlow D, Yang J, Clarke J, Dattani A, Yanagida A, Li MA, Myers S, Özel BN, Nichols J, Smith A. Human naive epiblast cells possess unrestricted lineage potential. Cell Stem Cell 2021; 28:1040-1056.e6. [PMID: 33831366 PMCID: PMC8189439 DOI: 10.1016/j.stem.2021.02.025] [Citation(s) in RCA: 216] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 09/17/2020] [Accepted: 02/23/2021] [Indexed: 01/04/2023]
Abstract
Classic embryological experiments have established that the early mouse embryo develops via sequential lineage bifurcations. The first segregated lineage is the trophectoderm, essential for blastocyst formation. Mouse naive epiblast and derivative embryonic stem cells are restricted accordingly from producing trophectoderm. Here we show, in contrast, that human naive embryonic stem cells readily make blastocyst trophectoderm and descendant trophoblast cell types. Trophectoderm was induced rapidly and efficiently by inhibition of ERK/mitogen-activated protein kinase (MAPK) and Nodal signaling. Transcriptome comparison with the human embryo substantiated direct formation of trophectoderm with subsequent differentiation into syncytiotrophoblast, cytotrophoblast, and downstream trophoblast stem cells. During pluripotency progression lineage potential switches from trophectoderm to amnion. Live-cell tracking revealed that epiblast cells in the human blastocyst are also able to produce trophectoderm. Thus, the paradigm of developmental specification coupled to lineage restriction does not apply to humans. Instead, epiblast plasticity and the potential for blastocyst regeneration are retained until implantation.
Collapse
Affiliation(s)
- Ge Guo
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK; Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK.
| | - Giuliano Giuseppe Stirparo
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK; Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK
| | - Stanley E Strawbridge
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - Daniel Spindlow
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - Jian Yang
- Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Sciences, Guangzhou 510530, China
| | - James Clarke
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - Anish Dattani
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK; Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK
| | - Ayaka Yanagida
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK; Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK
| | - Meng Amy Li
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - Sam Myers
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - Buse Nurten Özel
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - Jennifer Nichols
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK; Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 1GA, UK.
| | - Austin Smith
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK; Department of Biochemistry, University of Cambridge, Cambridge CB2 1QR, UK; Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK.
| |
Collapse
|
20
|
Kinoshita M, Barber M, Mansfield W, Cui Y, Spindlow D, Stirparo GG, Dietmann S, Nichols J, Smith A. Capture of Mouse and Human Stem Cells with Features of Formative Pluripotency. Cell Stem Cell 2021; 28:453-471.e8. [PMID: 33271069 PMCID: PMC7939546 DOI: 10.1016/j.stem.2020.11.005] [Citation(s) in RCA: 147] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 09/03/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023]
Abstract
Pluripotent cells emerge as a naive founder population in the blastocyst, acquire capacity for germline and soma formation, and then undergo lineage priming. Mouse embryonic stem cells (ESCs) and epiblast-derived stem cells (EpiSCs) represent the initial naive and final primed phases of pluripotency, respectively. Here, we investigate the intermediate formative stage. Using minimal exposure to specification cues, we derive stem cells from formative mouse epiblast. Unlike ESCs or EpiSCs, formative stem (FS) cells respond directly to germ cell induction. They colonize somatic tissues and germline in chimeras. Whole-transcriptome analyses show similarity to pre-gastrulation formative epiblast. Signal responsiveness and chromatin accessibility features reflect lineage capacitation. Furthermore, FS cells show distinct transcription factor dependencies, relying critically on Otx2. Finally, FS cell culture conditions applied to human naive cells or embryos support expansion of similar stem cells, consistent with a conserved staging post on the trajectory of mammalian pluripotency.
Collapse
Affiliation(s)
- Masaki Kinoshita
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK.
| | - Michael Barber
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - William Mansfield
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - Yingzhi Cui
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, UK
| | - Daniel Spindlow
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK; Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK
| | - Giuliano Giuseppe Stirparo
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK; Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK
| | - Sabine Dietmann
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - Jennifer Nichols
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK; Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | - Austin Smith
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK; Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, UK; Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK.
| |
Collapse
|
21
|
FOXC1 Downregulates Nanog Expression by Recruiting HDAC2 to Its Promoter in F9 Cells Treated by Retinoic Acid. Int J Mol Sci 2021; 22:ijms22052255. [PMID: 33668324 PMCID: PMC7956269 DOI: 10.3390/ijms22052255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/16/2021] [Accepted: 02/20/2021] [Indexed: 12/12/2022] Open
Abstract
FOXC1, a transcription factor involved in cell differentiation and embryogenesis, is demonstrated to be a negative regulator of Nanog in this study. FOXC1 is up-regulated in retinoic acid-induced differentiation of F9 Embryonal Carcinoma (EC) cells; furthermore, FOXC1 specifically inhibits the core pluripotency factor Nanog by binding to the proximal promoter. Overexpression of FOXC1 in F9 or knockdown in 3T3 results in the down-regulation or up-regulation of Nanog mRNA and proteins, respectively. In order to explain the mechanism by which FOXC1 inhibits Nanog expression, we identified the co-repressor HDAC2 from the FOXC1 interactome. FOXC1 recruits HDAC2 to Nanog promoter to decrease H3K27ac enrichment, resulting in transcription inhibition of Nanog. To the best of our knowledge, this is the first report that FOXC1 is involved in the epigenetic regulation of gene expression.
Collapse
|
22
|
Wnt/Beta-catenin/Esrrb signalling controls the tissue-scale reorganization and maintenance of the pluripotent lineage during murine embryonic diapause. Nat Commun 2020; 11:5499. [PMID: 33127892 PMCID: PMC7603494 DOI: 10.1038/s41467-020-19353-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 10/02/2020] [Indexed: 12/12/2022] Open
Abstract
The epiblast, which provides the foundation of the future body, is actively reshaped during early embryogenesis, but the reshaping mechanisms are poorly understood. Here, using a 3D in vitro model of early epiblast development, we identify the canonical Wnt/β-catenin pathway and its central downstream factor Esrrb as the key signalling cascade regulating the tissue-scale organization of the murine pluripotent lineage. Although in vivo the Wnt/β-catenin/Esrrb circuit is dispensable for embryonic development before implantation, autocrine Wnt activity controls the morphogenesis and long-term maintenance of the epiblast when development is put on hold during diapause. During this phase, the progressive changes in the epiblast architecture and Wnt signalling response show that diapause is not a stasis but instead is a dynamic process with underlying mechanisms that can appear redundant during transient embryogenesis. Embryonic diapause is a state of dormancy with poorly understood mechanisms of embryo intrinsic regulation. Here, the authors show that murine diapause is a dynamic process, where tissue-scale reorganization of the pluripotent lineage is controlled in an autocrine manner by the Wnt/b-catenin/Esrrb signalling cascade.
Collapse
|
23
|
Saiz N, Hadjantonakis AK. Coordination between patterning and morphogenesis ensures robustness during mouse development. Philos Trans R Soc Lond B Biol Sci 2020; 375:20190562. [PMID: 32829684 PMCID: PMC7482220 DOI: 10.1098/rstb.2019.0562] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2020] [Indexed: 12/11/2022] Open
Abstract
The mammalian preimplantation embryo is a highly tractable, self-organizing developmental system in which three cell types are consistently specified without the need for maternal factors or external signals. Studies in the mouse over the past decades have greatly improved our understanding of the cues that trigger symmetry breaking in the embryo, the transcription factors that control lineage specification and commitment, and the mechanical forces that drive morphogenesis and inform cell fate decisions. These studies have also uncovered how these multiple inputs are integrated to allocate the right number of cells to each lineage despite inherent biological noise, and as a response to perturbations. In this review, we summarize our current understanding of how these processes are coordinated to ensure a robust and precise developmental outcome during early mouse development. This article is part of a discussion meeting issue 'Contemporary morphogenesis'.
Collapse
Affiliation(s)
- Néstor Saiz
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | | |
Collapse
|
24
|
The Key Role of MicroRNAs in Self-Renewal and Differentiation of Embryonic Stem Cells. Int J Mol Sci 2020; 21:ijms21176285. [PMID: 32877989 PMCID: PMC7504502 DOI: 10.3390/ijms21176285] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/21/2020] [Accepted: 08/28/2020] [Indexed: 12/17/2022] Open
Abstract
Naïve pluripotent embryonic stem cells (ESCs) and epiblast stem cells (EpiSCs) represent distinctive developmental stages, mimicking the pre- and the post-implantation events during the embryo development, respectively. The complex molecular mechanisms governing the transition from ESCs into EpiSCs are orchestrated by fluctuating levels of pluripotency transcription factors (Nanog, Oct4, etc.) and wide-ranging remodeling of the epigenetic landscape. Recent studies highlighted the pivotal role of microRNAs (miRNAs) in balancing the switch from self-renewal to differentiation of ESCs. Of note, evidence deriving from miRNA-based reprogramming strategies underscores the role of the non-coding RNAs in the induction and maintenance of the stemness properties. In this review, we revised recent studies concerning the functions mediated by miRNAs in ESCs, with the aim of giving a comprehensive view of the highly dynamic miRNA-mediated tuning, essential to guarantee cell cycle progression, pluripotency maintenance and the proper commitment of ESCs.
Collapse
|
25
|
Alba G, Martínez R, Postigo-Corrales F, López S, Santa-María C, Jiménez J, Cahuana GM, Soria B, Bedoya FJ, Tejedo JR. AICAR Stimulates the Pluripotency Transcriptional Complex in Embryonic Stem Cells Mediated by PI3K, GSK3β, and β-Catenin. ACS OMEGA 2020; 5:20270-20282. [PMID: 32832780 PMCID: PMC7439381 DOI: 10.1021/acsomega.0c02137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/23/2020] [Indexed: 05/03/2023]
Abstract
Pluripotent stem cells maintain the property of self-renewal and differentiate into all cell types under clear environments. Though the gene regulatory mechanism for pluripotency has been investigated in recent years, it is still not completely understood. Here, we show several signaling pathways involved in the maintenance of pluripotency. To investigate whether AMPK is involved in maintaining the pluripotency in mouse embryonic stem cells (mESCs) and elucidating the possible molecular mechanisms, implicated D3 and R1/E mESC lines were used in this study. Cells were cultured in the absence or presence of LIF and treated with 1 mM and 0.5 mM 5-aminoimidazole-4-carboxamide-1-β-d-ribofuranoside (AICAR), 2 mM metformin, compound C, and the PI3K inhibitor LY294002 for 24, 72, and 120 h. The levels of Nanog, Oct3/4, and REX1 and Brachyury, Notch2, and Gata4 mRNAs and Nanog or OCT3/4 protein levels were analyzed. Alkaline phosphatase and the cellular cycle were determined. The pGSK3β, GSK3β, p-β-catenin, and β-catenin protein levels were also investigated. We found that AMPK activators such as AICAR and metformin increase mRNA expression of pluripotency markers and decrease mRNA expression of differentiation markers in R1/E and D3 ES cells. AICAR increases phosphatase activity and arrests the cellular cycle in the G1 phase in these cells. We describe that AICAR effects were mediated by AMPK activation using a chemical inhibitor or by silencing this gene. AICAR effects were also mediated by PI3K, GSK3β, and β-catenin in R1/E ES cells. According to our findings, we provide a mechanism by which AICAR increases and maintains a pluripotency state through enhanced Nanog expression, involving AMPK/PI3K and p-GSK3β Ser21/9 pathways backing up the AICAR function as a potential target for this drug controlling pluripotency. The highlights of this study are that AICAR (5-aminoimidazole-4-carboxamied-1-b-riboside), an AMP protein kinase (AMPK) activator, blocks the ESC differentiation and AMPK is a key enzyme for pluripotency and shows valuable data to clarify the molecular pluripotency mechanism.
Collapse
Affiliation(s)
- Gonzalo Alba
- Department
of Medical Biochemistry and Molecular Biology, Universidad de Sevilla, Seville 41009, Spain
- . Telephone: +34-955421044. Fax: +34-954907048
| | - Raquel Martínez
- Department
of Regeneration and Cell Therapy, Andalusian Center for Molecular
Biology and Regenerative Medicine-CABIMER, Universidad Pablo de Olavide-University of Seville-CSIC, Seville 41013, Spain
| | - Fátima Postigo-Corrales
- Department
of Regeneration and Cell Therapy, Andalusian Center for Molecular
Biology and Regenerative Medicine-CABIMER, Universidad Pablo de Olavide-University of Seville-CSIC, Seville 41013, Spain
| | - Soledad López
- Department
of Medical Biochemistry and Molecular Biology, Universidad de Sevilla, Seville 41009, Spain
| | - Consuelo Santa-María
- Department
of Biochemistry and Molecular Biology, Universidad
de Sevilla, Seville 41009, Spain
| | - Juan Jiménez
- Department
of Medical Biochemistry and Molecular Biology, Universidad de Sevilla, Seville 41009, Spain
| | - Gladys M. Cahuana
- Department
of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, Seville 41013, Spain
- Biomedical
Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM,
Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Bernat Soria
- Department
of Regeneration and Cell Therapy, Andalusian Center for Molecular
Biology and Regenerative Medicine-CABIMER, Universidad Pablo de Olavide-University of Seville-CSIC, Seville 41013, Spain
- Biomedical
Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM,
Instituto de Salud Carlos III, Madrid 28029, Spain
- Cell
Therapy
Network, Madrid (RED-TERCEL), Instituto
de Salud Carlos III, Madrid 28029, Spain
- Universidad
Miguel Hernández, Alicante 03550, Spain
| | - Francisco J. Bedoya
- Department
of Regeneration and Cell Therapy, Andalusian Center for Molecular
Biology and Regenerative Medicine-CABIMER, Universidad Pablo de Olavide-University of Seville-CSIC, Seville 41013, Spain
- Department
of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, Seville 41013, Spain
- Biomedical
Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM,
Instituto de Salud Carlos III, Madrid 28029, Spain
- Cell
Therapy
Network, Madrid (RED-TERCEL), Instituto
de Salud Carlos III, Madrid 28029, Spain
| | - Juan R. Tejedo
- Department
of Regeneration and Cell Therapy, Andalusian Center for Molecular
Biology and Regenerative Medicine-CABIMER, Universidad Pablo de Olavide-University of Seville-CSIC, Seville 41013, Spain
- Department
of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, Seville 41013, Spain
- Biomedical
Research Network for Diabetes and Related Metabolic Diseases-CIBERDEM,
Instituto de Salud Carlos III, Madrid 28029, Spain
- Cell
Therapy
Network, Madrid (RED-TERCEL), Instituto
de Salud Carlos III, Madrid 28029, Spain
| |
Collapse
|
26
|
Saiz N, Mora-Bitria L, Rahman S, George H, Herder JP, Garcia-Ojalvo J, Hadjantonakis AK. Growth-factor-mediated coupling between lineage size and cell fate choice underlies robustness of mammalian development. eLife 2020; 9:e56079. [PMID: 32720894 PMCID: PMC7513828 DOI: 10.7554/elife.56079] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 07/24/2020] [Indexed: 01/03/2023] Open
Abstract
Precise control and maintenance of population size is fundamental for organismal development and homeostasis. The three cell types of the mammalian blastocyst are generated in precise proportions over a short time, suggesting a mechanism to ensure a reproducible outcome. We developed a minimal mathematical model demonstrating growth factor signaling is sufficient to guarantee this robustness and which anticipates an embryo's response to perturbations in lineage composition. Addition of lineage-restricted cells both in vivo and in silico, causes a shift of the fate of progenitors away from the supernumerary cell type, while eliminating cells using laser ablation biases the specification of progenitors toward the targeted cell type. Finally, FGF4 couples fate decisions to lineage composition through changes in local growth factor concentration, providing a basis for the regulative abilities of the early mammalian embryo whereby fate decisions are coordinated at the population level to robustly generate tissues in the right proportions.
Collapse
Affiliation(s)
- Néstor Saiz
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Laura Mora-Bitria
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona Biomedical Research Park, Barcelona, Spain
| | - Shahadat Rahman
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Hannah George
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Jeremy P Herder
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Jordi Garcia-Ojalvo
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona Biomedical Research Park, Barcelona, Spain
| | - Anna-Katerina Hadjantonakis
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, United States
| |
Collapse
|
27
|
Sousa MI, Correia B, Rodrigues AS, Ramalho-Santos J. Metabolic characterization of a paused-like pluripotent state. Biochim Biophys Acta Gen Subj 2020; 1864:129612. [PMID: 32272203 DOI: 10.1016/j.bbagen.2020.129612] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 03/02/2020] [Accepted: 04/03/2020] [Indexed: 01/07/2023]
Abstract
Embryonic diapause is a conserved reproductive strategy in which development arrests at the blastocyst phase. Recently mammalian target of rapamycin (mTOR) inhibition was shown to induce diapause on mouse blastocysts and a paused-like state on mouse embryonic stem cells (mESCs). In this work, we aimed to further characterize this new paused-pluripotent state, focusing on its glycolytic and oxidative metabolic function. We therefore exposed mESCs, to the mTOR inhibitor INK-128 and evaluated proliferation, pluripotency status and energy-related metabolism, as well as the mTOR inhibition status and translational function. Unexpectedly, in our hands INK-128 did not inhibit the phosphorylation of mTOR or its downstream targets after 48 h. Accordingly, no alterations on protein translational function were observed. Nonetheless, INK-128 could still successfully induce a paused-like state in naïve mESCs regardless of their culturing conditions, by greatly slowing proliferation without affecting pluripotency status. This effect was more prevalent in 2i cultured cells. Interestingly, in this paused-like state, mESCs present a glucose-related hypometabolic profile, which is a hallmark of diapaused blastocysts, with decreased glycolytic and oxidative metabolism and decreased nutrient uptake. Despite the lack of mTOR inhibition and translational suppression, INK-128 still induced a paused-like pluripotent state through cell cycle and metabolic modulation, rather than by translational suppression, suggesting more than one avenue for this type of pluripotent phenotype.
Collapse
Affiliation(s)
- Maria Inês Sousa
- CNC-Center for Neuroscience and Cell Biology, CIBB, Azinhaga de Santa Comba, Polo 3, University of Coimbra, Coimbra, Portugal; University of Coimbra, Department of Life Sciences, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal
| | - Bibiana Correia
- CNC-Center for Neuroscience and Cell Biology, CIBB, Azinhaga de Santa Comba, Polo 3, University of Coimbra, Coimbra, Portugal
| | - Ana Sofia Rodrigues
- CNC-Center for Neuroscience and Cell Biology, CIBB, Azinhaga de Santa Comba, Polo 3, University of Coimbra, Coimbra, Portugal.
| | - João Ramalho-Santos
- CNC-Center for Neuroscience and Cell Biology, CIBB, Azinhaga de Santa Comba, Polo 3, University of Coimbra, Coimbra, Portugal; University of Coimbra, Department of Life Sciences, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal.
| |
Collapse
|
28
|
Bai X, Xu J, Zhu T, He Y, Zhang H. The Development of Stem Cell-Based Treatment for Acute Ischemic Cerebral Injury. Curr Stem Cell Res Ther 2020; 15:509-521. [PMID: 32228429 DOI: 10.2174/1574888x15666200331135227] [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: 12/19/2019] [Revised: 02/11/2020] [Accepted: 03/30/2020] [Indexed: 11/22/2022]
Abstract
Acute ischemic brain injury is a serious disease that severely endangers the life safety of patients. Such disease is hard to predict and highly lethal with very limited effective treatments currently. Although currently, there exist treatments like drug therapy, hyperbaric oxygen therapy, rehabilitation therapy and other treatments in clinical practice, these are not significantly effective for patients when the situation is severe. Thus scientists must explore more effective treatments. Stem cells are undifferentiated cells with a strong potential of self-renewal and differentiate into various types of tissues and organs. Their emergence has brought new hopes for overcoming difficult diseases, further improving medical technology and promoting the development of modern medicine. Some combining therapies and genetically modified stem cell therapy have also been proven to produce obvious neuroprotective function for acute ischemic brain injury. This review is an introduction to the current research findings and discusses the definition, origin and classification of stem cells, as well as the future prospects of the stem cell-based treatment for acute ischemic cerebral injury.
Collapse
Affiliation(s)
- Xiaojie Bai
- Department of Emergency, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Jun Xu
- Department of Emergency, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Tiantian Zhu
- Department of Emergency, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Yuanyuan He
- Department of Emergency, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Hong Zhang
- Department of Emergency, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| |
Collapse
|
29
|
Gomez-Salazar M, Gonzalez-Galofre ZN, Casamitjana J, Crisan M, James AW, Péault B. Five Decades Later, Are Mesenchymal Stem Cells Still Relevant? Front Bioeng Biotechnol 2020; 8:148. [PMID: 32185170 PMCID: PMC7058632 DOI: 10.3389/fbioe.2020.00148] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 02/13/2020] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells are culture-derived mesodermal progenitors isolatable from all vascularized tissues. In spite of multiple fundamental, pre-clinical and clinical studies, the native identity and role in tissue repair of MSCs have long remained elusive, with MSC selection in vitro from total cell suspensions essentially unchanged as a mere primary culture for half a century. Recent investigations have helped understand the tissue origin of these progenitor cells, and uncover alternative effects of MSCs on tissue healing via growth factor secretion and interaction with the immune system. In this review, we describe current trends in MSC biology and discuss how these may improve the use of these therapeutic cells in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Mario Gomez-Salazar
- MRC Centre for Regenerative Medicine and Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom
| | - Zaniah N Gonzalez-Galofre
- MRC Centre for Regenerative Medicine and Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom
| | - Joan Casamitjana
- MRC Centre for Regenerative Medicine and Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom
| | - Mihaela Crisan
- MRC Centre for Regenerative Medicine and Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom
| | - Aaron W James
- Orthopaedic Hospital Research Center and Broad Stem Cell Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Bruno Péault
- MRC Centre for Regenerative Medicine and Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, United Kingdom.,Orthopaedic Hospital Research Center and Broad Stem Cell Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
30
|
Samanta M, Kalantry S. Generating primed pluripotent epiblast stem cells: A methodology chapter. Curr Top Dev Biol 2020; 138:139-174. [PMID: 32220296 DOI: 10.1016/bs.ctdb.2020.01.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
At least two distinct pluripotent states, referred to as naïve and primed, define the early mammalian embryo. In the mouse, the pluripotent epiblast cells in the pre/peri-implantation embryo are the source of naïve embryonic stem cells (ESCs). After the embryo implants, the epiblast lineage generates a restricted or primed population of stem cells, referred to as epiblast stem cells (EpiSCs). ESCs can be cultured in EpiSC media to generate epiblast-like cells (EpiLCs). The differentiation of naive ESCs into primed EpiLCs permits insights into the development and differentiation of the pluripotent epiblast lineage. This chapter describes the generation and characterization of EpiSCs as well as EpiLCs.
Collapse
Affiliation(s)
- Milan Samanta
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Sundeep Kalantry
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, United States.
| |
Collapse
|
31
|
GP130 signaling and the control of naïve pluripotency in humans, monkeys, and pigs. Exp Cell Res 2020; 386:111712. [DOI: 10.1016/j.yexcr.2019.111712] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 10/31/2019] [Accepted: 11/02/2019] [Indexed: 12/19/2022]
|
32
|
Geng T, Zhang D, Jiang W. Epigenetic Regulation of Transition Among Different Pluripotent States: Concise Review. Stem Cells 2019; 37:1372-1380. [PMID: 31339608 DOI: 10.1002/stem.3064] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 06/12/2019] [Accepted: 07/08/2019] [Indexed: 12/11/2022]
Abstract
The extraordinary progress of pluripotent stem cell research provides a revolutionary avenue to understand mammalian early embryonic development. Besides well-established conventional mouse and human embryonic stem cells, the discoveries of naive state human stem cell, two-cell-like cell, and the newly defined "extended pluripotent" stem cell and "expanded potential" stem cell with bidirectional chimeric ability have greatly broadened the horizons of more pluripotent states recaptured and maintained in dish, infinitely approaching the totipotent blastomere state. Although all these pluripotent cell types can self-renew and have the ability to differentiate into all the three germ layers, accumulating evidence suggests that these pluripotent states display distinct epigenetic characters. More strikingly, epigenetic reprogramming, including DNA methylation, histone modification, and chromatin remodeling, is required to reset the cell fate commitment, suggesting that epigenetic mechanisms may play an active and important role in the maintenance and transition among these pluripotent states. Here, we have reviewed studies on various pluripotent states, with a highlight on the epigenetic regulation during the interconversion. Stem Cells 2019;37:1372-1380.
Collapse
Affiliation(s)
- Ting Geng
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University; Medical Research Institute, Wuhan University; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Hubei Province, People's Republic of China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, Hubei Province, People's Republic of China
| | - Wei Jiang
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University; Medical Research Institute, Wuhan University; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Hubei Province, People's Republic of China
| |
Collapse
|
33
|
Abbey D, Singh G, Verma I, Derebail S, Kolkundkar U, Chandrashekar DS, Acharya KK, Vemuri MC, Seshagiri PB. Successful Derivation of an Induced Pluripotent Stem Cell Line from a Genetically Nonpermissive Enhanced Green Fluorescent Protein-Transgenic FVB/N Mouse Strain. Cell Reprogram 2019; 21:270-284. [PMID: 31596624 DOI: 10.1089/cell.2019.0019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The embryonic stem cell line derivation from nonpermissive mouse strains is a challenging and highly inefficient process. The cellular reprogramming strategy provides an alternative route for generating pluripotent stem cell (PSC) lines from such strains. In this study, we successfully derived an enhanced green fluorescent protein (EGFP)-transgenic "N9" induced pluripotent stem cell (iPS cell, iPSC) line from the FVB/N strain-derived mouse embryonic fibroblasts (MEFs). The exposure of MEFs to human OCT4, SOX2, KLF4, and c-MYC (OSKM) transgenes via lentiviral transduction resulted in complete reprogramming. The N9 iPS cell line demonstrated all the criteria of a typical mouse PSC line, including normal colony morphology and karyotype (40,XY), high replication and propagation efficiencies, expression of the pluripotency-associated genes, spontaneous differentiation to three germ lineage-derived cell types, and robust potential of chimeric blastocyst formation. Taken together, using human OSKM genes for transduction, we report, for the first time, the successful derivation of an EGFP-expressing iPS cell line from a genetically nonpermissive transgenic FVB/N mouse. This cell line could provide opportunities for designing protocols for efficient derivation of PSC lines from other nonpermissive strains and developing mouse models of human diseases.
Collapse
Affiliation(s)
- Deepti Abbey
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| | - Gurbind Singh
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India.,Present address: Centre for Stem Cell Research, Christian Medical College Campus, Bagayam, Vellore, India
| | - Isha Verma
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| | | | | | | | | | | | - Polani B Seshagiri
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| |
Collapse
|
34
|
Harvey A, Caretti G, Moresi V, Renzini A, Adamo S. Interplay between Metabolites and the Epigenome in Regulating Embryonic and Adult Stem Cell Potency and Maintenance. Stem Cell Reports 2019; 13:573-589. [PMID: 31597110 PMCID: PMC6830055 DOI: 10.1016/j.stemcr.2019.09.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/29/2019] [Accepted: 09/06/2019] [Indexed: 12/15/2022] Open
Abstract
The environment surrounding stem cells has the ability to elicit profound, heritable epigenetic changes orchestrated by multiple epigenetic mechanisms, which can be modulated by the level of specific metabolites. In this review, we highlight the significance of metabolism in regulating stem cell homeostasis, cell state, and differentiation capacity, using metabolic regulation of embryonic and adult muscle stem cells as examples, and cast light on the interaction between cellular metabolism and epigenetics. These new regulatory networks, based on the dynamic interplay between metabolism and epigenetics in stem cell biology, are important, not only for understanding tissue homeostasis, but to determine in vitro culture conditions which accurately support normal cell physiology.
Collapse
Affiliation(s)
- Alexandra Harvey
- School of BioSciences, University of Melbourne, Parkville, VIC 2010, Australia
| | - Giuseppina Caretti
- Department of Biosciences, Università degli Studi di Milano, Milan, Italy
| | - Viviana Moresi
- Department of Anatomy, Histology, Forensic Medicine & Orthopedics, Histology & Medical Embryology Section, Sapienza University of Rome and Interuniversity Institute of Myology, Rome, Italy.
| | - Alessandra Renzini
- Department of Anatomy, Histology, Forensic Medicine & Orthopedics, Histology & Medical Embryology Section, Sapienza University of Rome and Interuniversity Institute of Myology, Rome, Italy
| | - Sergio Adamo
- Department of Anatomy, Histology, Forensic Medicine & Orthopedics, Histology & Medical Embryology Section, Sapienza University of Rome and Interuniversity Institute of Myology, Rome, Italy
| |
Collapse
|
35
|
Fiorenza MT, Rava A. The TCL1 function revisited focusing on metabolic requirements of stemness. Cell Cycle 2019; 18:3055-3063. [PMID: 31564197 DOI: 10.1080/15384101.2019.1672465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The oncogenic ability of the T-cell leukemia/lymphoma 1 gene, TCL1, has captured the attention in the field of prolymphocytic T-cell and B-cell chronic leukemias for more than two decades. However, the finding that TCL1 is also expressed in totipotent cells of the mouse preimplantation embryos and that it is among the 10 genes, including the transcription factors Nanog, Oct4, Sox2, Tbx3, and Esrrb, that are required for maintaining the mitotic self-renewal state of embryonic stem cells, raises a great interest. In this review, we highlight newly acquired evidence pinpointing TCL1 as a crucial regulator of metabolic pathways that dictate somatic cell reprogramming toward pluripotency. In our opinion, this feature provides a relevant hint for reframing the role that this factor plays at early stages of mammalian embryo development and in tumorigenesis. Hence, the TCL1-dependent enhancement of serine/threonine AKT/PKB kinase activity favoring cell proliferation appears to be associated to the promotion of glucose transport and activation of glycolytic pathways. This is also consistent with the TCL1 ability to suppress mitochondrial biogenesis and oxygen consumption, downplaying the contribution of oxidative phosphorylation to energy metabolism. It thus appears that TCL1 masters the direction of energy metabolism toward the glycolytic pathway to meet a critical metabolic requirement that goes beyond the mere ATP production. For instance, the synthesis of glycolytic intermediates that are required for DNA synthesis likely represents the most pressing cellular need for both cleavage-stage embryos and rapidly proliferating tumor cells.
Collapse
Affiliation(s)
- Maria Teresa Fiorenza
- Department of Psychology, Division of Neuroscience and "Daniel Bovet" Neurobiology Research Center, Sapienza University of Rome , Rome , Italy.,IRCCS Fondazione Santa Lucia , Rome , Italy
| | - Alessandro Rava
- Department of Psychology, Division of Neuroscience and "Daniel Bovet" Neurobiology Research Center, Sapienza University of Rome , Rome , Italy
| |
Collapse
|
36
|
Wang Y, Guo B, Xiao Z, Lin H, Zhang X, Song Y, Li Y, Gao X, Yu J, Shao Z, Li X, Luo Y, Li S. Long noncoding RNA CCDC144NL-AS1 knockdown induces naïve-like state conversion of human pluripotent stem cells. Stem Cell Res Ther 2019; 10:220. [PMID: 31358062 PMCID: PMC6664583 DOI: 10.1186/s13287-019-1323-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 06/18/2019] [Accepted: 07/02/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Human naïve pluripotency state cells can be derived from direct isolation of inner cell mass or primed-to-naïve resetting of human embryonic stem cells (hESCs) through different combinations of transcription factors, small molecular inhibitors, and growth factors. Long noncoding RNAs (lncRNAs) have been identified to be crucial in diverse biological processes, including pluripotency regulatory circuit of mouse pluripotent stem cells (PSCs), but few are involved in human PSCs' regulation of pluripotency and naïve pluripotency derivation. This study initially planned to discover more lncRNAs possibly playing significant roles in the regulation of human PSCs' pluripotency, but accidently identified a lncRNA whose knockdown in human PSCs induced naïve-like pluripotency conversion. METHODS Candidate lncRNAs tightly correlated with human pluripotency were screened from 55 RNA-seq data containing human ESC, human induced pluripotent stem cell (iPSC), and somatic tissue samples. Then loss-of-function experiments in human PSCs were performed to investigate the function of these candidate lncRNAs. The naïve-like pluripotency conversion caused by CCDC144NL-AS1 knockdown (KD) was characterized by quantitative real-time PCR, immunofluorescence staining, western blotting, differentiation of hESCs in vitro and in vivo, RNA-seq, and chromatin immunoprecipitation. Finally, the signaling pathways in CCDC144NL-AS1-KD human PSCs were examined through western blotting and analysis of RNA-seq data. RESULTS The results indicated that knockdown of CCDC144NL-AS1 induces naïve-like state conversion of human PSCs in the absence of additional transcription factors or small molecular inhibitors. CCDC144NL-AS1-KD human PSCs reveal naïve-like pluripotency features, such as elevated expression of naïve pluripotency-associated genes, increased developmental capacity, analogous transcriptional profiles to human naïve PSCs, and global reduction of repressive chromatin modification marks. Furthermore, CCDC144NL-AS1-KD human PSCs display inhibition of MAPK (ERK), accumulation of active β-catenin, and upregulation of some LIF/STAT3 target genes, and all of these are concordant with previously reported traits of human naïve PSCs. CONCLUSIONS Our study unveils an unexpected role of a lncRNA, CCDC144NL-AS1, in the naïve-like state conversion of human PSCs, providing a new perspective to further understand the regulation process of human early pluripotency states conversion. It is suggested that CCDC144NL-AS1 can be potentially valuable for future research on deriving higher quality naïve state human PSCs and promoting their therapeutic applications.
Collapse
Affiliation(s)
- Yingying Wang
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Baosen Guo
- College of Life Sciences, Nanchang University, Nanchang, 330031, China
| | - Zengrong Xiao
- College of Life Sciences, Nanchang University, Nanchang, 330031, China
| | - Haijun Lin
- College of Life Sciences, Nanchang University, Nanchang, 330031, China
| | - Xi Zhang
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Yueqiang Song
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Yalei Li
- College of Life Sciences, Nanchang University, Nanchang, 330031, China
| | - Xuehu Gao
- College of Life Sciences, Nanchang University, Nanchang, 330031, China
| | - Jinjun Yu
- College of Life Sciences, Nanchang University, Nanchang, 330031, China
| | - Zhihua Shao
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Xuekun Li
- The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310052, China
| | - Yuping Luo
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China. .,Human Aging Research Institute and School of Life Science, Nanchang University, Nanchang, 330031, China.
| | - Siguang Li
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China. .,Collaborative Innovation Center for Brain Science, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
37
|
Zhang M, Leitch HG, Tang WWC, Festuccia N, Hall-Ponsele E, Nichols J, Surani MA, Smith A, Chambers I. Esrrb Complementation Rescues Development of Nanog-Null Germ Cells. Cell Rep 2019; 22:332-339. [PMID: 29320730 PMCID: PMC5775501 DOI: 10.1016/j.celrep.2017.12.060] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 11/15/2017] [Accepted: 12/17/2017] [Indexed: 12/24/2022] Open
Abstract
The transcription factors (TFs) Nanog and Esrrb play important roles in embryonic stem cells (ESCs) and during primordial germ-cell (PGC) development. Esrrb is a positively regulated direct target of NANOG in ESCs that can substitute qualitatively for Nanog function in ESCs. Whether this functional substitution extends to the germline is unknown. Here, we show that germline deletion of Nanog reduces PGC numbers 5-fold at midgestation. Despite this quantitative depletion, Nanog-null PGCs can complete germline development in contrast to previous findings. PGC-like cell (PGCLC) differentiation of Nanog-null ESCs is also impaired, with Nanog-null PGCLCs showing decreased proliferation and increased apoptosis. However, induced expression of Esrrb restores PGCLC numbers as efficiently as Nanog. These effects are recapitulated in vivo: knockin of Esrrb to Nanog restores PGC numbers to wild-type levels and results in fertile adult mice. These findings demonstrate that Esrrb can replace Nanog function in germ cells. Germline deletion of Nanog reduces PGC numbers but does not abolish PGC development Without Nanog, PGCLCs form ineffectively with less proliferation and more apoptosis The Nanog target gene Esrrb can rescue PGCLC differentiation of Nanog−/− ESCs Knockin of Esrrb at the Nanog locus restores PGC development efficiency
Collapse
Affiliation(s)
- Man Zhang
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, Scotland
| | - Harry G Leitch
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, United Kingdom.
| | - Walfred W C Tang
- Wellcome Trust/Cancer Research UK Gurdon Institute, Tennis Court Road, University of Cambridge, Cambridge CB2 1QN, United Kingdom; Department of Physiology, Development and Neuroscience, Downing Street, University of Cambridge, Cambridge CB2 3EG, United Kingdom
| | - Nicola Festuccia
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, Scotland
| | - Elisa Hall-Ponsele
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, Scotland
| | - Jennifer Nichols
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, United Kingdom; Department of Physiology, Development and Neuroscience, Downing Street, University of Cambridge, Cambridge CB2 3EG, United Kingdom
| | - M Azim Surani
- Wellcome Trust/Cancer Research UK Gurdon Institute, Tennis Court Road, University of Cambridge, Cambridge CB2 1QN, United Kingdom; Department of Physiology, Development and Neuroscience, Downing Street, University of Cambridge, Cambridge CB2 3EG, United Kingdom
| | - Austin Smith
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, United Kingdom; Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, United Kingdom
| | - Ian Chambers
- MRC Centre for Regenerative Medicine, Institute for Stem Cell Research, School of Biological Sciences, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, Scotland.
| |
Collapse
|
38
|
Rostovskaya M, Stirparo GG, Smith A. Capacitation of human naïve pluripotent stem cells for multi-lineage differentiation. Development 2019; 146:dev172916. [PMID: 30944104 PMCID: PMC6467473 DOI: 10.1242/dev.172916] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 02/22/2019] [Indexed: 12/14/2022]
Abstract
Human naïve pluripotent stem cells (PSCs) share features with the pre-implantation epiblast. They therefore provide an unmatched opportunity for characterising the developmental programme of pluripotency in Homo sapiens Here, we confirm that naïve PSCs do not respond directly to germ layer induction, but must first acquire competence. Capacitation for multi-lineage differentiation occurs without exogenous growth factor stimulation and is facilitated by inhibition of Wnt signalling. Whole-transcriptome profiling during this formative transition highlights dynamic changes in gene expression, which affect many cellular properties including metabolism and epithelial features. Notably, naïve pluripotency factors are exchanged for postimplantation factors, but competent cells remain devoid of lineage-specific transcription. The gradual pace of transition for human naïve PSCs is consistent with the timespan of primate development from blastocyst to gastrulation. Transcriptome trajectory during in vitro capacitation of human naïve cells tracks the progression of the epiblast during embryogenesis in Macaca fascicularis, but shows greater divergence from mouse development. Thus, the formative transition of naïve PSCs in a simple culture system may recapitulate essential and specific features of pluripotency dynamics during an inaccessible period of human embryogenesis.
Collapse
Affiliation(s)
- Maria Rostovskaya
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge CB2 1QR, United Kingdom
| | - Giuliano G Stirparo
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge CB2 1QR, United Kingdom
| | - Austin Smith
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge CB2 1QR, United Kingdom
| |
Collapse
|
39
|
Mulas C, Kalkan T, von Meyenn F, Leitch HG, Nichols J, Smith A. Defined conditions for propagation and manipulation of mouse embryonic stem cells. Development 2019; 146:dev173146. [PMID: 30914406 PMCID: PMC6451320 DOI: 10.1242/dev.173146] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 02/19/2019] [Indexed: 02/02/2023]
Abstract
The power of mouse embryonic stem (ES) cells to colonise the developing embryo has revolutionised mammalian developmental genetics and stem cell research. This power is vulnerable, however, to the cell culture environment, deficiencies in which can lead to cellular heterogeneity, adaptive phenotypes, epigenetic aberrations and genetic abnormalities. Here, we provide detailed methodologies for derivation, propagation, genetic modification and primary differentiation of ES cells in 2i or 2i+LIF media without serum or undefined serum substitutes. Implemented diligently, these procedures minimise variability and deviation, thereby improving the efficiency, reproducibility and biological validity of ES cell experimentation.
Collapse
Affiliation(s)
- Carla Mulas
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Tüzer Kalkan
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Ferdinand von Meyenn
- Department of Medical and Molecular Genetics, King's College London, London SE1 9RT, UK
| | - Harry G Leitch
- MRC London Institute of Medical Sciences (LMS), Du Cane Road, London W12 0NN, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Jennifer Nichols
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3DY, UK
| | - Austin Smith
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
- Department of Biochemistry, University of Cambridge, Hopkins Building, Tennis Court Road, Cambridge CB2 1QW, UK
| |
Collapse
|
40
|
Hassani SN, Moradi S, Taleahmad S, Braun T, Baharvand H. Transition of inner cell mass to embryonic stem cells: mechanisms, facts, and hypotheses. Cell Mol Life Sci 2019; 76:873-892. [PMID: 30420999 PMCID: PMC11105545 DOI: 10.1007/s00018-018-2965-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 11/01/2018] [Accepted: 11/02/2018] [Indexed: 12/28/2022]
Abstract
Embryonic stem cells (ESCs) are immortal stem cells that own multi-lineage differentiation potential. ESCs are commonly derived from the inner cell mass (ICM) of pre-implantation embryos. Due to their tremendous developmental capacity and unlimited self-renewal, ESCs have diverse biomedical applications. Different culture media have been developed to procure and maintain ESCs in a state of naïve pluripotency, and to preserve a stable genome and epigenome during serial passaging. Chromatin modifications such as DNA methylation and histone modifications along with microRNA activity and different signaling pathways dynamically contribute to the regulation of the ESC gene regulatory network (GRN). Such modifications undergo remarkable changes in different ESC media and determine the quality and developmental potential of ESCs. In this review, we discuss the current approaches for derivation and maintenance of ESCs, and examine how differences in culture media impact on the characteristics of pluripotency via modulation of GRN during the course of ICM outgrowth into ESCs. We also summarize the current hypotheses concerning the origin of ESCs and provide a perspective about the relationship of these cells to their in vivo counterparts (early embryonic cells around the time of implantation). Finally, we discuss generation of ESCs from human embryos and domesticated animals, and offer suggestions to further advance this fascinating field.
Collapse
Affiliation(s)
- Seyedeh-Nafiseh Hassani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Sharif Moradi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Sara Taleahmad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Thomas Braun
- Department of Cardiac Development and Remodelling, Max-Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran.
| |
Collapse
|
41
|
Vila-Cejudo M, Massafret O, Santaló J, Ibáñez E. Single blastomeres as a source of mouse embryonic stem cells: effect of genetic background, medium supplements, and signaling modulators on derivation efficiency. J Assist Reprod Genet 2019; 36:99-111. [PMID: 30430313 PMCID: PMC6338609 DOI: 10.1007/s10815-018-1360-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 10/30/2018] [Indexed: 11/24/2022] Open
Abstract
PURPOSE To assess the role of the genetic background, the culture medium supplements, and the presence of modulators of signaling pathways on mouse embryonic stem cell derivation from single blastomeres from 8-cell embryos. METHODS Mice from permissive and non-permissive genetic backgrounds, different culture media supplements, knockout serum replacement (KSR) and N2B27, and the presence or absence of 2i treatment were used to derive mouse embryonic stem cells (mESC) from single blastomeres isolated from 8-cell embryos and from control embryos at the blastocyst stage. After the sixth passage, the putative mESC were analyzed by immunofluorescence to assess their pluripotency and, after in vitro differentiation induction, their ability to differentiate into derivatives of the three primary germ layers. Selected mESC lines derived from single blastomeres in the most efficient culture conditions were further characterized to validate their stemness. RESULTS In control embryos, high mESC derivation efficiencies (70-96.9%) were obtained from permissive backgrounds or when embryos were cultured in medium complemented with 2i regardless of their genetic background. By contrast, only blastomeres isolated from embryos from permissive background cultured in KSR-containing medium complemented with 2i were moderately successful in the derivation of mESC lines (22.9-24.5%). Moreover, we report for the first time that B6CBAF2 embryos behave as permissive in terms of mESC derivation. CONCLUSIONS Single blastomeres have higher requirements than whole blastocysts for pluripotency maintenance and mESC derivation. The need for 2i suggests that modulation of signaling pathways to recreate a commitment towards inner cell mass could be essential to efficiently derive mESC from single blastomeres.
Collapse
Affiliation(s)
- Marta Vila-Cejudo
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Tissue Engineering Unit, Centre for Genomic Regulation, Barcelona, Spain
| | - Ot Massafret
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Josep Santaló
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Elena Ibáñez
- Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain.
| |
Collapse
|
42
|
Afanassieff M, Aksoy I, Beaujean N, Bourillot PY, Savatier P. [Fifty shades of pluripotency]. Med Sci (Paris) 2018; 34:944-953. [PMID: 30526839 DOI: 10.1051/medsci/2018240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Since the derivation of the first pluripotent embryonic stem cell lines in mice in the early 1980s, a plethora of lines has been obtained from various mammalian species including rodents, lagomorphs and primates. These lines are distinguished by their molecular and functional characteristics and correspond to the different pluripotency states observed in the developing embryo between the "blastocyst" and "gastrula" stages. These cell lines are positioned along a gradient, or continuum of pluripotency, the ends of which are epitomized by the naïve and primed states, respectively. Conventional human pluripotent stem cells self-renew in the primed state of pluripotency (ie, at the bottom of the gradient), a position that is undoubtedly the cause of their natural instability. Recent studies aim to generate naive human pluripotent stem cells (at the top of the gradient). The importance of this research in the perspective of medical applications will be discussed.
Collapse
Affiliation(s)
- Marielle Afanassieff
- Univ Lyon, Université Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, INRA USC 1361, 69500 Bron, France
| | - Irène Aksoy
- Univ Lyon, Université Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, INRA USC 1361, 69500 Bron, France
| | - Nathalie Beaujean
- Univ Lyon, Université Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, INRA USC 1361, 69500 Bron, France
| | - Pierre-Yves Bourillot
- Univ Lyon, Université Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, INRA USC 1361, 69500 Bron, France
| | - Pierre Savatier
- Univ Lyon, Université Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, INRA USC 1361, 69500 Bron, France
| |
Collapse
|
43
|
Plusa B, Hadjantonakis AK. (De)constructing the blastocyst: Lessons in self-organization from the mouse. ACTA ACUST UNITED AC 2018. [DOI: 10.1016/j.coisb.2018.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
44
|
Lager AM, Corradin OG, Cregg JM, Elitt MS, Shick HE, Clayton BLL, Allan KC, Olsen HE, Madhavan M, Tesar PJ. Rapid functional genetics of the oligodendrocyte lineage using pluripotent stem cells. Nat Commun 2018; 9:3708. [PMID: 30213958 PMCID: PMC6137209 DOI: 10.1038/s41467-018-06102-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 08/14/2018] [Indexed: 01/08/2023] Open
Abstract
Oligodendrocyte dysfunction underlies many neurological disorders, but rapid assessment of mutation-specific effects in these cells has been impractical. To enable functional genetics in oligodendrocytes, here we report a highly efficient method for generating oligodendrocytes and their progenitors from mouse embryonic and induced pluripotent stem cells, independent of mouse strain or mutational status. We demonstrate that this approach, when combined with genome engineering, provides a powerful platform for the expeditious study of genotype-phenotype relationships in oligodendrocytes.
Collapse
Affiliation(s)
- Angela M Lager
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Olivia G Corradin
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Jared M Cregg
- Department of Neurosciences, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Matthew S Elitt
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
| | - H Elizabeth Shick
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Benjamin L L Clayton
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Kevin C Allan
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Hannah E Olsen
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Mayur Madhavan
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH, 44106, USA
| | - Paul J Tesar
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH, 44106, USA. .,Department of Neurosciences, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, OH, 44106, USA.
| |
Collapse
|
45
|
Jafari N, Giehr P, Hesaraki M, Baas R, de Graaf P, Timmers HTM, Walter J, Baharvand H, Totonchi M. Genomic integrity of ground-state pluripotency. J Cell Biochem 2018; 119:9781-9789. [PMID: 30171711 DOI: 10.1002/jcb.27296] [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: 12/17/2017] [Accepted: 06/25/2018] [Indexed: 11/06/2022]
Abstract
Pluripotent cells appear to be in a transient state during early development. These cells have the capability to transition into embryonic stem cells (ESCs). It has been reported that mouse pluripotent cells cultivated in chemically defined media sustain the ground state of pluripotency. Because the epigenetic pattern of pluripotent cells reflects their environment, culture under different conditions causes epigenetic changes, which could lead to genomic instability. This study focused on the DNA methylation pattern of repetitive elements (REs) and their activation levels under two ground-state conditions and assessed the genomic integrity of ESCs. We measured the methylation and expression level of REs in different media. The results indicated that although the ground-state conditions show higher REs activity, they did not lead to DNA damage; therefore, the level of genomic instability is lower under the ground-state compared with the conventional condition. Our results indicated that when choosing an optimum condition, different features of the condition must be considered to have epigenetically and genomically stable stem cells.
Collapse
Affiliation(s)
- Narges Jafari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Pascal Giehr
- Department of Genetics/Epigenetics, Saarland University, Saarbrücken, Germany
| | - Mahdi Hesaraki
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Roy Baas
- Regenerative Medicine Center and Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Petra de Graaf
- Regenerative Medicine Center and Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - H T Marc Timmers
- Regenerative Medicine Center and Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Urology, German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (DKFZ), University Medical Center Freiburg, Freiburg, Germany
| | - Jörn Walter
- Department of Genetics/Epigenetics, Saarland University, Saarbrücken, Germany
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Mehdi Totonchi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
46
|
Nett IR, Mulas C, Gatto L, Lilley KS, Smith A. Negative feedback via RSK modulates Erk-dependent progression from naïve pluripotency. EMBO Rep 2018; 19:e45642. [PMID: 29895711 PMCID: PMC6073214 DOI: 10.15252/embr.201745642] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 05/16/2018] [Accepted: 05/18/2018] [Indexed: 01/08/2023] Open
Abstract
Mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signalling is implicated in initiation of embryonic stem (ES) cell differentiation. The pathway is subject to complex feedback regulation. Here, we examined the ERK-responsive phosphoproteome in ES cells and identified the negative regulator RSK1 as a prominent target. We used CRISPR/Cas9 to create combinatorial mutations in RSK family genes. Genotypes that included homozygous null mutations in Rps6ka1, encoding RSK1, resulted in elevated ERK phosphorylation. These RSK-depleted ES cells exhibit altered kinetics of transition into differentiation, with accelerated downregulation of naïve pluripotency factors, precocious expression of transitional epiblast markers and early onset of lineage specification. We further show that chemical inhibition of RSK increases ERK phosphorylation and expedites ES cell transition without compromising multilineage potential. These findings demonstrate that the ERK activation profile influences the dynamics of pluripotency progression and highlight the role of signalling feedback in temporal control of cell state transitions.
Collapse
Affiliation(s)
- Isabelle Re Nett
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Carla Mulas
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Laurent Gatto
- Department of Biochemistry, Cambridge Centre for Proteomics, University of Cambridge, Cambridge, UK
- Computational Proteomics Unit, Department of Biochemistry, Cambridge Centre for Proteomics, University of Cambridge, Cambridge, UK
| | - Kathryn S Lilley
- Department of Biochemistry, Cambridge Centre for Proteomics, University of Cambridge, Cambridge, UK
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Austin Smith
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
47
|
Du Y, Wang T, Xu J, Zhao C, Li H, Fu Y, Xu Y, Xie L, Zhao J, Yang W, Yin M, Wen J, Deng H. Efficient derivation of extended pluripotent stem cells from NOD-scid Il2rg -/- mice. Protein Cell 2018; 10:31-42. [PMID: 29948854 PMCID: PMC6321811 DOI: 10.1007/s13238-018-0558-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 05/16/2018] [Indexed: 11/11/2022] Open
Abstract
Recently we have established a new culture condition enabling the derivation of extended pluripotent stem (EPS) cells, which, compared to conventional pluripotent stem cells, possess superior developmental potential and germline competence. However, it remains unclear whether this condition permits derivation of EPS cells from mouse strains that are refractory or non-permissive to pluripotent cell establishment. Here, we show that EPS cells can be robustly generated from non-permissive NOD-scid Il2rg−/− mice through de novo derivation from blastocysts. Furthermore, these cells can also be efficiently generated by chemical reprogramming from embryonic NOD-scid Il2rg−/− fibroblasts. NOD-scid Il2rg−/− EPS cells can be expanded for more than 20 passages with genomic stability and can be genetically modified through gene targeting. Notably, these cells contribute to both embryonic and extraembryonic lineages in vivo. More importantly, they can produce chimeras and integrate into the E13.5 genital ridge. Our study demonstrates the feasibility of generating EPS cells from refractory mouse strains, which could potentially be a general strategy for deriving mouse pluripotent cells. The generation of NOD-scid Il2rg−/− EPS cell lines permits sophisticated genetic modification in NOD-scid Il2rg−/− mice, which may greatly advance the optimization of humanized mouse models for biomedical applications.
Collapse
Affiliation(s)
- Yaqin Du
- Peking University Stem Cell Research Center, Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Ting Wang
- Peking University Stem Cell Research Center, Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Jun Xu
- Peking University Stem Cell Research Center, Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Chaoran Zhao
- Peking University Stem Cell Research Center, Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Haibo Li
- Peking University Stem Cell Research Center, Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yao Fu
- The MOE Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Yaxing Xu
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Liangfu Xie
- The MOE Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Jingru Zhao
- Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Weifeng Yang
- Beijing Vitalstar Biotechnology, Beijing, 100012, China
| | - Ming Yin
- Beijing Vitalstar Biotechnology, Beijing, 100012, China
| | - Jinhua Wen
- Peking University Stem Cell Research Center, Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| | - Hongkui Deng
- Peking University Stem Cell Research Center, Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China. .,Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, College of Life Sciences, Peking University, Beijing, 100871, China. .,The MOE Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
48
|
Talbot NC, Caperna TJ, Willard RR, Meekin JH, Garrett WM. Characterization of Two Subpopulations of the PICM-19 Porcine Liver Stem Cell Line for use in Cell-Based Extracorporeal Liver Assistance Devices. Int J Artif Organs 2018. [DOI: 10.1177/039139881003300603] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Two cell lines, PICM-19H and PICM-19B, were derived from the bipotent PICM-19 pig liver stem cell line and assessed for their potential application in artificial liver devices (ALD). The study included assessments of growth rate and cell density in culture, morphological features, serum protein production, γ-glutamyltranspeptidase (GGT) activity and hepatocyte detoxification functions, i.e., inducible P450 activity, ammonia clearance, and urea production. The PICM-19H cell line was derived by temperature selection at 33–34°C. After each passage, PICM-19H cells grew to a nearly confluent monolayer of cells of hepatocyte morphology, i.e., cuboidal cells with centrally located nuclei joined by biliary canaliculi. No differentiation and self-organization into multi-cellular bile ductules, as observed in the parental PICM-19 cell line, occurred within the PICM-19H cell monolayers. The PICM-19H cells contained numerous mitochondria, Golgi apparatus, smooth and rough endoplasmic reticulum, vesicular bodies and occasional lipid vacuoles. The cells had a doubling time of 48–72 h and reached a final density of 1.5 x 105 cells/cm2 at ∼10 d post-passage from a 1:6 split ratio. PICM-19H cells displayed inducible P450 activity, cleared ammonia, and produced urea in a glutamine-free medium. The PICM-19B cells were colony-cloned after spontaneous generation from the PICM-19 parental cell line. PICM-19B cells grew as a tightly knit dome-forming monolayer with no visible biliary canaliculi. Their doubling time was 48–72 h with a final cell density of 2.6 x 105 cells/cm2. Ultrastructural analysis of the PICM-19B monolayers showed the roughly cuboidal cells displayed basal-apical polarization and were joined by tight junction-like complexes. Other ultrastructure features were similar to those of PICM-19H cells except that they possessed numerous cell bodies resembling mucus vacuoles. The PICM-19B cells had relatively high levels of GGT activity, but did retain some inducible P450 activity, and some ammonia clearance and urea synthesis ability. PICM-19B cells produced markedly less serum proteins than PICM-19H cells. These data indicated that both cell lines, either together or alone, may be useful as the cellular substrate for an ALD.
Collapse
Affiliation(s)
- Neil C. Talbot
- US Department of Agriculture, Agricultural Research Service, Animal and Natural Resources Institute, Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, Beltsville, Maryland - USA
| | - Thomas J. Caperna
- US Department of Agriculture, Agricultural Research Service, Animal and Natural Resources Institute, Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, Beltsville, Maryland - USA
| | - Ryan R. Willard
- US Department of Agriculture, Agricultural Research Service, Animal and Natural Resources Institute, Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, Beltsville, Maryland - USA
| | - John H. Meekin
- HepaLife Technologies, Inc., Boston, Massachusetts - USA
| | - Wesley M. Garrett
- US Department of Agriculture, Agricultural Research Service, Animal and Natural Resources Institute, Animal Biosciences and Biotechnology Laboratory, Beltsville Agricultural Research Center, Beltsville, Maryland - USA
| |
Collapse
|
49
|
Efficient derivation of stable primed pluripotent embryonic stem cells from bovine blastocysts. Proc Natl Acad Sci U S A 2018; 115:2090-2095. [PMID: 29440377 DOI: 10.1073/pnas.1716161115] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Embryonic stem cells (ESCs) are derived from the inner cell mass of preimplantation blastocysts. From agricultural and biomedical perspectives, the derivation of stable ESCs from domestic ungulates is important for genomic testing and selection, genome engineering, and modeling human diseases. Cattle are one of the most important domestic ungulates that are commonly used for food and bioreactors. To date, however, it remains a challenge to produce stable pluripotent bovine ESC lines. Employing a culture system containing fibroblast growth factor 2 and an inhibitor of the canonical Wnt-signaling pathway, we derived pluripotent bovine ESCs (bESCs) with stable morphology, transcriptome, karyotype, population-doubling time, pluripotency marker gene expression, and epigenetic features. Under this condition bESC lines were efficiently derived (100% in optimal conditions), were established quickly (3-4 wk), and were simple to propagate (by trypsin treatment). When used as donors for nuclear transfer, bESCs produced normal blastocyst rates, thereby opening the possibility for genomic selection, genome editing, and production of cattle with high genetic value.
Collapse
|
50
|
From Flies to Mice: The Emerging Role of Non-Canonical PRC1 Members in Mammalian Development. EPIGENOMES 2018. [DOI: 10.3390/epigenomes2010004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|