1
|
Slamecka J, Ryu S, Tristan CA, Chu PH, Weber C, Deng T, Gedik Y, Ormanoglu P, Voss TC, Simeonov A, Singeç I. Highly efficient generation of self-renewing trophoblast from human pluripotent stem cells. iScience 2024; 27:110874. [PMID: 39386760 PMCID: PMC11462042 DOI: 10.1016/j.isci.2024.110874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 06/02/2024] [Accepted: 08/30/2024] [Indexed: 10/12/2024] Open
Abstract
Human pluripotent stem cells (hPSCs) represent a powerful model system to study early developmental processes. However, lineage specification into trophectoderm (TE) and trophoblast (TB) differentiation remains poorly understood, and access to well-characterized placental cells for biomedical research is limited, largely depending on fetal tissues or cancer cell lines. Here, we developed novel strategies enabling highly efficient TE specification that generates cytotrophoblast (CTB) and multinucleated syncytiotrophoblast (STB), followed by the establishment of trophoblast stem cells (TSCs) capable of differentiating into extravillous trophoblast (EVT) and STB after long-term expansion. We confirmed stepwise and controlled induction of lineage- and cell-type-specific genes consistent with developmental biology principles and benchmarked typical features of placental cells using morphological, biochemical, genomics, epigenomics, and single-cell analyses. Charting a well-defined roadmap from hPSCs to distinct placental phenotypes provides invaluable opportunities for studying early human development, infertility, and pregnancy-associated diseases.
Collapse
Affiliation(s)
- Jaroslav Slamecka
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Seungmi Ryu
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Carlos A. Tristan
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Pei-Hsuan Chu
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Claire Weber
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Tao Deng
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Yeliz Gedik
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Pinar Ormanoglu
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Ty C. Voss
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Anton Simeonov
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Ilyas Singeç
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| |
Collapse
|
2
|
Lemke KA, Sarkar CA, Azarin SM. Rapid retinoic acid-induced trophoblast cell model from human induced pluripotent stem cells. Sci Rep 2024; 14:18204. [PMID: 39107470 PMCID: PMC11303561 DOI: 10.1038/s41598-024-68952-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
A limited number of accessible and representative models of human trophoblast cells currently exist for the study of placentation. Current stem cell models involve either a transition through a naïve stem cell state or precise dynamic control of multiple growth factors and small-molecule cues. Here, we demonstrated that a simple five-day treatment of human induced pluripotent stem cells with two small molecules, retinoic acid (RA) and Wnt agonist CHIR 99021 (CHIR), resulted in rapid, synergistic upregulation of CDX2. Transcriptomic analysis of RA + CHIR-treated cells showed high similarity to primary trophectoderm cells. Multipotency was verified via further differentiation towards cells with syncytiotrophoblast or extravillous trophoblast features. RA + CHIR-treated cells were also assessed for the established criteria defining a trophoblast cell model, and they possess all the features necessary to be considered valid. Collectively, our data demonstrate a facile, scalable method for generating functional trophoblast-like cells in vitro to better understand the placenta.
Collapse
Affiliation(s)
- Kristen A Lemke
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Casim A Sarkar
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Samira M Azarin
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
3
|
Shannon MJ, McNeill GL, Koksal B, Baltayeva J, Wächter J, Castellana B, Peñaherrera MS, Robinson WP, Leung PCK, Beristain AG. Single-cell assessment of primary and stem cell-derived human trophoblast organoids as placenta-modeling platforms. Dev Cell 2024; 59:776-792.e11. [PMID: 38359834 DOI: 10.1016/j.devcel.2024.01.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 11/15/2023] [Accepted: 01/23/2024] [Indexed: 02/17/2024]
Abstract
Human trophoblast stem cells (hTSCs) and related trophoblast organoids are state-of-the-art culture systems that facilitate the study of trophoblast development and human placentation. Using single-cell transcriptomics, we evaluate how organoids derived from freshly isolated first-trimester trophoblasts or from established hTSC cell lines reproduce developmental cell trajectories and transcriptional regulatory processes defined in vivo. Although organoids from primary trophoblasts and hTSCs overall model trophoblast differentiation with accuracy, specific features related to trophoblast composition, trophoblast differentiation, and transcriptional drivers of trophoblast development show levels of misalignment. This is best illustrated by the identification of an expanded progenitor state in stem cell-derived organoids that is nearly absent in vivo and transcriptionally shares both villous cytotrophoblast and extravillous trophoblast characteristics. Together, this work provides a comprehensive resource that identifies strengths and limitations of current trophoblast organoid platforms.
Collapse
Affiliation(s)
- Matthew J Shannon
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| | - Gina L McNeill
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| | - Burak Koksal
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| | - Jennet Baltayeva
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| | - Jasmin Wächter
- Department of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - Barbara Castellana
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| | - Maria S Peñaherrera
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Medical Genetics, The University of British Columbia, Vancouver, BC, Canada
| | - Wendy P Robinson
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Medical Genetics, The University of British Columbia, Vancouver, BC, Canada
| | - Peter C K Leung
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| | - Alexander G Beristain
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
4
|
Anvar Z, Chakchouk I, Sharif M, Mahadevan S, Su L, Anikar S, Naini FA, Utama AB, Van den Veyver IB. Comparison of Four Protocols for In Vitro Differentiation of Human Embryonic Stem Cells into Trophoblast Lineages by BMP4 and Dual Inhibition of Activin/Nodal and FGF2 Signaling. Reprod Sci 2024; 31:173-189. [PMID: 37658178 PMCID: PMC10784360 DOI: 10.1007/s43032-023-01334-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 08/16/2023] [Indexed: 09/03/2023]
Abstract
Human embryonic stem cells (hESCs) cultured in media containing bone morphogenic protein 4 (BMP4; B) differentiate into trophoblast-like cells. Supplementing media with inhibitors of activin/nodal signaling (A83-01) and of fibroblast growth factor 2 (PD173074) suppresses mesoderm and endoderm formation and improves specification of trophoblast-like lineages, but with variable effectiveness. We compared differentiation in four BMP4-containing media: mTeSR1-BMP4 only, mTeSR1-BAP, basal medium with BAP (basal-BAP), and a newly defined medium, E7-BAP. These media variably drive early differentiation towards trophoblast-like lineages with upregulation of early trophoblast markers CDX2 and KRT7 and downregulation of pluripotency markers (OCT4 and NANOG). As expected, based on differences between media in FGF2 and its inhibitors, downregulation of mesendoderm marker EOMES was variable between media. By day 7, only hESCs grown in E7-BAP or basal-BAP expressed HLA-G protein, indicating the presence of cells with extravillous trophoblast characteristics. Expression of HLA-G and other differentiation markers (hCG, KRT7, and GCM1) was highest in basal-BAP, suggesting a faster differentiation in this medium, but those cultures were more inhomogeneous and still expressed some endodermal and pluripotency markers. In E7-BAP, HLA-G expression increased later and was lower. There was also a low but maintained expression of some C19MC miRNAs, with more CpG hypomethylation of the ELF5 promoter, suggesting that E7-BAP cultures differentiate slower along the trophoblast lineage. We conclude that while all protocols drive differentiation into trophoblast lineages with varying efficiency, they have advantages and disadvantages that must be considered when selecting a protocol for specific experiments.
Collapse
Affiliation(s)
- Zahra Anvar
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
- Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Room 1025.14, Houston, TX, 77030, USA
| | - Imen Chakchouk
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
- Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Room 1025.14, Houston, TX, 77030, USA
| | - Momal Sharif
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
- Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Room 1025.14, Houston, TX, 77030, USA
| | - Sangeetha Mahadevan
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
- Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Room 1025.14, Houston, TX, 77030, USA
| | - Li Su
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
- Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Room 1025.14, Houston, TX, 77030, USA
| | - Swathi Anikar
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
- Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Room 1025.14, Houston, TX, 77030, USA
| | - Fatemeh Alavi Naini
- Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Room 1025.14, Houston, TX, 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | | | - Ignatia B Van den Veyver
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA.
- Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Room 1025.14, Houston, TX, 77030, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
5
|
Sadovsky E, Chu T, Barak O, Sadovsky Y, Ouyang Y. The impact of opioids on the transcriptional landscape of human villous trophoblasts. Placenta 2023; 143:54-61. [PMID: 37832183 PMCID: PMC10841529 DOI: 10.1016/j.placenta.2023.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 09/15/2023] [Accepted: 10/05/2023] [Indexed: 10/15/2023]
Abstract
INTRODUCTION Opioid use disorder (OUD) is implicated in major obstetrical diseases such as fetal growth restriction. Whether or not opioids directly impact placental trophoblast development and function remains unclear. We sought to examine the expression of opioid receptors (OPRs) in villous trophoblasts and the effect of opioids on placental transcriptomics. METHODS Trophoblast stem (TS) cells and primary human trophoblast (PHT) cells from healthy term placentas were used to assess OPR expression in conditions that enhance trophoblast stemness vs differentiation. Placental RNAseq was conducted using our retrospective cohorts of pregnant people with OUD vs controls, both without major obstetrical complications. RT-qPCR was used to determine the effect of fentanyl on the expression of putative opioid targets and stemness or differentiation-associated genes in TS and PHT cells. RESULTS Three main OPRs, including OPRM1, OPRD1, and OPRK1 were expressed in term PHT cells cultured in the stemness medium, whereas only OPRD1 and OPRK1 were expressed in TS cells. Interestingly, upon induction of differentiation, the expressed OPR mRNAs in TS or in PHT cells were downregulated. We found 286 differentially expressed long RNAs in placentas from the OUD participants vs controls. While three putative opioid targets differed their expression in stemness vs differentiation states of trophoblasts, fentanyl had no effect on their expression or the expression of major stemness or differentiation-relevant genes in TS and PHT cells. DISCUSSION Trophoblastic expression of OPRs and opioid RNA targets is impacted by cell differentiation, suggesting differential susceptibility of villous trophoblasts to the effect of opioids.
Collapse
Affiliation(s)
- Elena Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tianjiao Chu
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Oren Barak
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yoel Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA; Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yingshi Ouyang
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
6
|
Huang CC, Hsueh YW, Chang CW, Hsu HC, Yang TC, Lin WC, Chang HM. Establishment of the fetal-maternal interface: developmental events in human implantation and placentation. Front Cell Dev Biol 2023; 11:1200330. [PMID: 37266451 PMCID: PMC10230101 DOI: 10.3389/fcell.2023.1200330] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/09/2023] [Indexed: 06/03/2023] Open
Abstract
Early pregnancy is a complex and well-orchestrated differentiation process that involves all the cellular elements of the fetal-maternal interface. Aberrant trophoblast-decidual interactions can lead to miscarriage and disorders that occur later in pregnancy, including preeclampsia, intrauterine fetal growth restriction, and preterm labor. A great deal of research on the regulation of implantation and placentation has been performed in a wide range of species. However, there is significant species variation regarding trophoblast differentiation as well as decidual-specific gene expression and regulation. Most of the relevant information has been obtained from studies using mouse models. A comprehensive understanding of the physiology and pathology of human implantation and placentation has only recently been obtained because of emerging advanced technologies. With the derivation of human trophoblast stem cells, 3D-organoid cultures, and single-cell analyses of differentiated cells, cell type-specific transcript profiles and functions were generated, and each exhibited a unique signature. Additionally, through integrative transcriptomic information, researchers can uncover the cellular dysfunction of embryonic and placental cells in peri-implantation embryos and the early pathological placenta. In fact, the clinical utility of fetal-maternal cellular trafficking has been applied for the noninvasive prenatal diagnosis of aneuploidies and the prediction of pregnancy complications. Furthermore, recent studies have proposed a viable path toward the development of therapeutic strategies targeting placenta-enriched molecules for placental dysfunction and diseases.
Collapse
|
7
|
Chen ACH, Lee YL, Ruan H, Huang W, Fong SW, Tian S, Lee KC, Wu GM, Tan Y, Wong TCH, Wu J, Zhang W, Cao D, Chow JFC, Liu P, Yeung WSB. Expanded Potential Stem Cells from Human Embryos Have an Open Chromatin Configuration with Enhanced Trophoblast Differentiation Ability. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204797. [PMID: 36775869 PMCID: PMC10104645 DOI: 10.1002/advs.202204797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 01/27/2023] [Indexed: 06/18/2023]
Abstract
Human expanded potential stem cells (hEPSC) have been derived from human embryonic stem cells and induced pluripotent stem cells. Here direct derivation of hEPSC from human pre-implantation embryos is reported. Like the reported hEPSC, the embryo-derived hEPSC (hEPSC-em) exhibit a transcriptome similar to morula, comparable differentiation potency, and high genome editing efficiency. Interestingly, the hEPSC-em show a unique H3 lysine-4 trimethylation (H3K4me3) open chromatin conformation; they possess a higher proportion of H3K4me3 bound broad domain (>5 kb) than the reported hEPSC, naive, and primed embryonic stem cells. The open conformation is associated with enhanced trophoblast differentiation potency with increased trophoblast gene expression upon induction of differentiation and success in derivation of trophoblast stem cells with bona fide characteristics. Hippo signaling is specifically enriched in the H3K4me3 broad domains of the hEPSC-. Knockout of the Hippo signaling gene, YAP1 abolishes the ability of the embryo-derived EPSC to form trophoblast stem cells.
Collapse
Affiliation(s)
- Andy Chun Hang Chen
- Department of Obstetrics and Gynaecology, School of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
- Shenzhen Key Laboratory of Fertility RegulationReproductive Medicine CenterThe University of Hong Kong ‐ Shenzhen HospitalShenzhen518000China
- Centre for Translational Stem Cell BiologyBuilding 17 WThe Hong Kong Science and Technology ParkHong KongHong Kong
| | - Yin Lau Lee
- Department of Obstetrics and Gynaecology, School of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
- Shenzhen Key Laboratory of Fertility RegulationReproductive Medicine CenterThe University of Hong Kong ‐ Shenzhen HospitalShenzhen518000China
- Centre for Translational Stem Cell BiologyBuilding 17 WThe Hong Kong Science and Technology ParkHong KongHong Kong
| | - Hanzhang Ruan
- Department of Obstetrics and Gynaecology, School of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
| | - Wen Huang
- Department of Obstetrics and Gynaecology, School of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
| | - Sze Wan Fong
- Department of Obstetrics and Gynaecology, School of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
| | - Siyu Tian
- Department of Obstetrics and Gynaecology, School of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
| | - Kai Chuen Lee
- Department of Obstetrics and Gynaecology, School of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
- Centre for Translational Stem Cell BiologyBuilding 17 WThe Hong Kong Science and Technology ParkHong KongHong Kong
| | - Genie Minju Wu
- Department of Obstetrics and Gynaecology, School of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
| | - Yongqi Tan
- Department of Obstetrics and Gynaecology, School of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
| | - Timothy Chun Hin Wong
- Centre for Translational Stem Cell BiologyBuilding 17 WThe Hong Kong Science and Technology ParkHong KongHong Kong
| | - Jian Wu
- Centre for Translational Stem Cell BiologyBuilding 17 WThe Hong Kong Science and Technology ParkHong KongHong Kong
| | - Weiyu Zhang
- Centre for Translational Stem Cell BiologyBuilding 17 WThe Hong Kong Science and Technology ParkHong KongHong Kong
| | - Dandan Cao
- Shenzhen Key Laboratory of Fertility RegulationReproductive Medicine CenterThe University of Hong Kong ‐ Shenzhen HospitalShenzhen518000China
| | - Judy Fung Cheung Chow
- Department of Obstetrics and Gynaecology, School of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
| | - Pengtao Liu
- Centre for Translational Stem Cell BiologyBuilding 17 WThe Hong Kong Science and Technology ParkHong KongHong Kong
- School of Biomedical SciencesLi Ka Shing Faculty of MedicineThe University of Hong KongStem Cell and Regenerative Medicine ConsortiumHong KongHong Kong
| | - William Shu Biu Yeung
- Department of Obstetrics and Gynaecology, School of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
- Shenzhen Key Laboratory of Fertility RegulationReproductive Medicine CenterThe University of Hong Kong ‐ Shenzhen HospitalShenzhen518000China
- Centre for Translational Stem Cell BiologyBuilding 17 WThe Hong Kong Science and Technology ParkHong KongHong Kong
| |
Collapse
|
8
|
Li H, Peng H, Hong W, Wei Y, Tian H, Huang X, Jia L, Zheng J, Duan T, He Q, Wang K. Human Placental Endothelial Cell and Trophoblast Heterogeneity and Differentiation Revealed by Single-Cell RNA Sequencing. Cells 2022; 12:cells12010087. [PMID: 36611882 PMCID: PMC9818681 DOI: 10.3390/cells12010087] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/21/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The placenta is an important organ for fetal and maternal health during pregnancy and impacts offspring health late in life. Defects in placental vasculature and trophoblast have been identified in several pregnancy complications. Thus, the detailed molecular profile and heterogeneity of endothelial cells and trophoblasts in placentas will aid us in better understanding placental behaviors and improving pregnancy outcomes. METHODS Single-cell RNA sequencing (scRNA-seq) was performed to profile the transcriptomics of human placental villous tissues from eleven patients with normal pregnancies in the first and second trimesters (6-16 weeks of gestation). RESULTS The transcriptomic landscape of 52,179 single cells was obtained, and the cells were classified as trophoblasts, fibroblasts, endothelial cells, erythroid cells, Hofbauer cells, and macrophages. Our analysis further revealed the three subtypes of placental endothelial cells, with distinct metabolic signatures and transcription factor regulatory networks. We also determined the transcriptomic features of the trophoblast subpopulations and characterized two distinct populations of progenitor cells in cytotrophoblasts, which were capable of differentiating to extravillous trophoblasts and syncytiotrophoblasts, respectively. CONCLUSIONS Our study provided a high-resolution molecular profile of the human placenta between 6 and 16 weeks of gestation. Our data revealed the placental cell complexity and demonstrated the transcriptional networks and signaling involved in placental endothelial and trophoblast differentiation during early pregnancy, which will be a resource for future studies of the human placental development.
Collapse
Affiliation(s)
- Han Li
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 201204, China
| | - Hao Peng
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 201204, China
| | - Wei Hong
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 201204, China
| | - Yingying Wei
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 201204, China
| | - Haojun Tian
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 201204, China
| | - Xiaojie Huang
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 201204, China
| | - Linyan Jia
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 201204, China
| | - Jing Zheng
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Tao Duan
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 201204, China
| | - Qizhi He
- Department of Pathology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 201204, China
- Correspondence: (Q.H.); (K.W.)
| | - Kai Wang
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 201204, China
- Correspondence: (Q.H.); (K.W.)
| |
Collapse
|
9
|
Karvas RM, David L, Theunissen TW. Accessing the human trophoblast stem cell state from pluripotent and somatic cells. Cell Mol Life Sci 2022; 79:604. [PMID: 36434136 PMCID: PMC9702929 DOI: 10.1007/s00018-022-04549-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/01/2022] [Accepted: 09/05/2022] [Indexed: 11/26/2022]
Abstract
Trophoblasts are specialized epithelial cells that perform critical functions during blastocyst implantation and mediate maternal-fetal communication during pregnancy. However, our understanding of human trophoblast biology remains limited since access to first-trimester placental tissue is scarce, especially between the first and fourth weeks of development. Moreover, animal models inadequately recapitulate unique aspects of human placental physiology. In the mouse system, the isolation of self-renewing trophoblast stem cells has provided a valuable in vitro model system of placental development, but the derivation of analogous human trophoblast stem cells (hTSCs) has remained elusive until recently. Building on a landmark study reporting the isolation of bona fide hTSCs from blastocysts and first-trimester placental tissues in 2018, several groups have developed methods to derive hTSCs from pluripotent and somatic cell sources. Here we review the biological and molecular properties that define authentic hTSCs, the trophoblast potential of distinct pluripotent states, and methods for inducing hTSCs in somatic cells by direct reprogramming. The generation of hTSCs from pluripotent and somatic cells presents exciting opportunities to elucidate the molecular mechanisms of human placental development and the etiology of pregnancy-related diseases.
Collapse
Affiliation(s)
- Rowan M Karvas
- Department of Developmental Biology and Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Laurent David
- Nantes Université, CHU Nantes, INSERM, CR2TI, UMR 1064, 44000, Nantes, France.
- Nantes Université, CHU Nantes, INSERM, CNRS, Biocore, US 016, UAR 3556, 44000, Nantes, France.
| | - Thorold W Theunissen
- Department of Developmental Biology and Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
10
|
Viukov S, Shani T, Bayerl J, Aguilera-Castrejon A, Oldak B, Sheban D, Tarazi S, Stelzer Y, Hanna JH, Novershtern N. Human primed and naïve PSCs are both able to differentiate into trophoblast stem cells. Stem Cell Reports 2022; 17:2484-2500. [PMID: 36270280 PMCID: PMC9669397 DOI: 10.1016/j.stemcr.2022.09.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 11/09/2022] Open
Abstract
The recent derivation of human trophoblast stem cells (TSCs) from placental cytotrophoblasts and blastocysts opened opportunities for studying the development and function of the human placenta. Recent reports have suggested that human naïve, but not primed, pluripotent stem cells (PSCs) retain an exclusive potential to generate TSCs. Here we report that, in the absence of WNT stimulation, transforming growth factor β (TGF-β) pathway inhibition leads to direct and robust conversion of primed human PSCs into TSCs. The resulting primed PSC-derived TSC lines exhibit self-renewal, can differentiate into the main trophoblast lineages, and present RNA and epigenetic profiles that are indistinguishable from recently established TSC lines derived from human placenta, blastocysts, or isogenic human naïve PSCs expanded under human enhanced naïve stem cell medium (HENSM) conditions. Activation of nuclear Yes-associated protein (YAP) signaling is sufficient for this conversion and necessary for human TSC maintenance. Our findings underscore a residual plasticity in primed human PSCs that allows their in vitro conversion into extra-embryonic trophoblast lineages. Primed human PSCs readily convert into TSCs upon inhibition of TGF-β pathway Human primed PSC-derived TSCs are similar to embryo- or naïve PSC-derived TSCs WNT activation inhibits conversion to TSC in primed but not in naïve hPSCs YAP is sufficient for TSC induction from hPSCs and necessary for TSC maintenance
Collapse
Affiliation(s)
- Sergey Viukov
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Tom Shani
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Jonathan Bayerl
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | | | - Bernardo Oldak
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Daoud Sheban
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Shadi Tarazi
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Yonatan Stelzer
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Jacob H Hanna
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel.
| | - Noa Novershtern
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
11
|
Ray S, Saha A, Ghosh A, Roy N, Kumar RP, Meinhardt G, Mukerjee A, Gunewardena S, Kumar R, Knöfler M, Paul S. Hippo signaling cofactor, WWTR1, at the crossroads of human trophoblast progenitor self-renewal and differentiation. Proc Natl Acad Sci U S A 2022; 119:e2204069119. [PMID: 36037374 PMCID: PMC9457323 DOI: 10.1073/pnas.2204069119] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 08/01/2022] [Indexed: 11/30/2022] Open
Abstract
Healthy progression of human pregnancy relies on cytotrophoblast (CTB) progenitor self-renewal and its differentiation toward multinucleated syncytiotrophoblasts (STBs) and invasive extravillous trophoblasts (EVTs). However, the underlying molecular mechanisms that fine-tune CTB self-renewal or direct its differentiation toward STBs or EVTs during human placentation are poorly defined. Here, we show that Hippo signaling cofactor WW domain containing transcription regulator 1 (WWTR1) is a master regulator of trophoblast fate choice during human placentation. Using human trophoblast stem cells (human TSCs), primary CTBs, and human placental explants, we demonstrate that WWTR1 promotes self-renewal in human CTBs and is essential for their differentiation to EVTs. In contrast, WWTR1 prevents induction of the STB fate in undifferentiated CTBs. Our single-cell RNA sequencing analyses in first-trimester human placenta, along with mechanistic analyses in human TSCs revealed that WWTR1 fine-tunes trophoblast fate by directly regulating WNT signaling components. Importantly, our analyses of placentae from pathological pregnancies show that extreme preterm births (gestational time ≤28 wk) are often associated with loss of WWTR1 expression in CTBs. In summary, our findings establish the critical importance of WWTR1 at the crossroads of human trophoblast progenitor self-renewal versus differentiation. It plays positive instructive roles in promoting CTB self-renewal and EVT differentiation and safeguards undifferentiated CTBs from attaining the STB fate.
Collapse
Affiliation(s)
- Soma Ray
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160
| | - Abhik Saha
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160
| | - Ananya Ghosh
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160
| | - Namrata Roy
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160
| | - Ram P. Kumar
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160
| | - Gudrun Meinhardt
- Department of Obstetrics and Gynecology, Reproductive Biology Unit, Placental Development Group, Medical University of Vienna, Vienna, Austria 1090
| | - Abhirup Mukerjee
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160
| | - Sumedha Gunewardena
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Rajnish Kumar
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160
| | - Martin Knöfler
- Department of Obstetrics and Gynecology, Reproductive Biology Unit, Placental Development Group, Medical University of Vienna, Vienna, Austria 1090
| | - Soumen Paul
- Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160
- Institute for Reproduction and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS 66160
| |
Collapse
|
12
|
Roberts RM, Ezashi T, Temple J, Owen JR, Soncin F, Parast MM. The role of BMP4 signaling in trophoblast emergence from pluripotency. Cell Mol Life Sci 2022; 79:447. [PMID: 35877048 PMCID: PMC10243463 DOI: 10.1007/s00018-022-04478-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/24/2022] [Accepted: 07/06/2022] [Indexed: 11/03/2022]
Abstract
The Bone Morphogenetic Protein (BMP) signaling pathway has established roles in early embryonic morphogenesis, particularly in the epiblast. More recently, however, it has also been implicated in development of extraembryonic lineages, including trophectoderm (TE), in both mouse and human. In this review, we will provide an overview of this signaling pathway, with a focus on BMP4, and its role in emergence and development of TE in both early mouse and human embryogenesis. Subsequently, we will build on these in vivo data and discuss the utility of BMP4-based protocols for in vitro conversion of primed vs. naïve pluripotent stem cells (PSC) into trophoblast, and specifically into trophoblast stem cells (TSC). PSC-derived TSC could provide an abundant, reproducible, and ethically acceptable source of cells for modeling placental development.
Collapse
Affiliation(s)
- R Michael Roberts
- Division of Animal Sciences and Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Toshihiko Ezashi
- Division of Animal Sciences and Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
- Colorado Center for Reproductive Medicine, 10290 Ridgegate Circle, Lone Tree, CO, 80124, USA
| | - Jasmine Temple
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA
| | - Joseph R Owen
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Francesca Soncin
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA
| | - Mana M Parast
- Department of Pathology, University of California San Diego, La Jolla, CA, USA.
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA.
| |
Collapse
|
13
|
Renaud SJ, Jeyarajah MJ. How trophoblasts fuse: an in-depth look into placental syncytiotrophoblast formation. Cell Mol Life Sci 2022; 79:433. [PMID: 35859055 PMCID: PMC11072895 DOI: 10.1007/s00018-022-04475-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/07/2022] [Accepted: 07/06/2022] [Indexed: 11/24/2022]
Abstract
In humans, cell fusion is restricted to only a few cell types under normal conditions. In the placenta, cell fusion is a critical process for generating syncytiotrophoblast: the giant multinucleated trophoblast lineage containing billions of nuclei within an interconnected cytoplasm that forms the primary interface separating maternal blood from fetal tissue. The unique morphology of syncytiotrophoblast ensures that nutrients and gases can be efficiently transferred between maternal and fetal tissue while simultaneously restricting entry of potentially damaging substances and maternal immune cells through intercellular junctions. To maintain integrity of the syncytiotrophoblast layer, underlying cytotrophoblast progenitor cells terminate their capability for self-renewal, upregulate expression of genes needed for differentiation, and then fuse into the overlying syncytium. These processes are disrupted in a variety of obstetric complications, underscoring the importance of proper syncytiotrophoblast formation for pregnancy health. Herein, an overview of key mechanisms underlying human trophoblast fusion and syncytiotrophoblast development is discussed.
Collapse
Affiliation(s)
- Stephen J Renaud
- Department of Anatomy and Cell Biology and Children's Health Research Institute, University of Western Ontario, London, ON, N6A5C1, Canada.
| | - Mariyan J Jeyarajah
- Department of Anatomy and Cell Biology and Children's Health Research Institute, University of Western Ontario, London, ON, N6A5C1, Canada
| |
Collapse
|
14
|
Single-cell transcriptional profiling reveals cellular and molecular divergence in human maternal-fetal interface. Sci Rep 2022; 12:10892. [PMID: 35764880 PMCID: PMC9240006 DOI: 10.1038/s41598-022-14516-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/08/2022] [Indexed: 12/19/2022] Open
Abstract
Placenta plays essential role in successful pregnancy, as the most important organ connecting and interplaying between mother and fetus. However, the cellular characteristics and molecular interaction of cell populations within the fetomaternal interface is still poorly understood. Here, we surveyed the single-cell transcriptomic landscape of human full-term placenta and revealed the heterogeneity of cytotrophoblast cell (CTB) and stromal cell (STR) with the fetal/maternal origin consecutively localized from fetal section (FS), middle section (Mid_S) to maternal section (Mat_S) of maternal–fetal interface. Then, we highlighted a subpopulation of CTB, named trophoblast progenitor-like cells (TPLCs) existed in the full-term placenta and mainly distributed in Mid_S, with high expression of a pool of putative cell surface markers. Further, we revealed the putative key transcription factor PRDM6 that might promote the differentiation of endovascular extravillous trophoblast cells (enEVT) by inhibiting cell proliferation, and down-regulation of PRDM6 might lead to an abnormal enEVT differentiation process in PE. Together, our study offers important resources for better understanding of human placenta and stem cell-based therapy, and provides new insights on the study of tissue heterogeneity, the clinical prevention and control of PE as well as the maternal–fetal interface.
Collapse
|
15
|
James JL, Lissaman A, Nursalim YNS, Chamley LW. Modelling human placental villous development: designing cultures that reflect anatomy. Cell Mol Life Sci 2022; 79:384. [PMID: 35753002 PMCID: PMC9234034 DOI: 10.1007/s00018-022-04407-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/12/2022] [Accepted: 05/30/2022] [Indexed: 11/03/2022]
Abstract
The use of in vitro tools to study trophoblast differentiation and function is essential to improve understanding of normal and abnormal placental development. The relative accessibility of human placentae enables the use of primary trophoblasts and placental explants in a range of in vitro systems. Recent advances in stem cell models, three-dimensional organoid cultures, and organ-on-a-chip systems have further shed light on the complex microenvironment and cell-cell crosstalk involved in placental development. However, understanding each model's strengths and limitations, and which in vivo aspects of human placentation in vitro data acquired does, or does not, accurately reflect, is key to interpret findings appropriately. To help researchers use and design anatomically accurate culture models, this review both outlines our current understanding of placental development, and critically considers the range of established and emerging culture models used to study this, with a focus on those derived from primary tissue.
Collapse
Affiliation(s)
- Joanna L James
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Abbey Lissaman
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Yohanes N S Nursalim
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Lawrence W Chamley
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
16
|
Soncin F, Morey R, Bui T, Requena DF, Cheung VC, Kallol S, Kittle R, Jackson MG, Farah O, Chousal J, Meads M, Pizzo D, Horii M, Fisch KM, Parast MM. Derivation of functional trophoblast stem cells from primed human pluripotent stem cells. Stem Cell Reports 2022; 17:1303-1317. [PMID: 35594858 PMCID: PMC9214048 DOI: 10.1016/j.stemcr.2022.04.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 12/25/2022] Open
Abstract
Trophoblast stem cells (TSCs) have recently been derived from human embryos and early-first-trimester placenta; however, aside from ethical challenges, the unknown disease potential of these cells limits their scientific utility. We have previously established a bone morphogetic protein 4 (BMP4)-based two-step protocol for differentiation of primed human pluripotent stem cells (hPSCs) into functional trophoblasts; however, those trophoblasts could not be maintained in a self-renewing TSC-like state. Here, we use the first step from this protocol, followed by a switch to newly developed TSC medium, to derive bona fide TSCs. We show that these cells resemble placenta- and naive hPSC-derived TSCs, based on their transcriptome as well as their in vitro and in vivo differentiation potential. We conclude that primed hPSCs can be used to generate functional TSCs through a simple protocol, which can be applied to a widely available set of existing hPSCs, including induced pluripotent stem cells, derived from patients with known birth outcomes.
Collapse
Affiliation(s)
- Francesca Soncin
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Robert Morey
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Tony Bui
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Daniela F Requena
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Virginia Chu Cheung
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Sampada Kallol
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Ryan Kittle
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Madeline G Jackson
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Omar Farah
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jennifer Chousal
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Morgan Meads
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Donald Pizzo
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA
| | - Mariko Horii
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Kathleen M Fisch
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Mana M Parast
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
17
|
Transcription factor networks in trophoblast development. Cell Mol Life Sci 2022; 79:337. [PMID: 35657505 PMCID: PMC9166831 DOI: 10.1007/s00018-022-04363-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 12/12/2022]
Abstract
The placenta sustains embryonic development and is critical for a successful pregnancy outcome. It provides the site of exchange between the mother and the embryo, has immunological functions and is a vital endocrine organ. To perform these diverse roles, the placenta comprises highly specialized trophoblast cell types, including syncytiotrophoblast and extravillous trophoblast. The coordinated actions of transcription factors (TFs) regulate their emergence during development, subsequent specialization, and identity. These TFs integrate diverse signaling cues, form TF networks, associate with chromatin remodeling and modifying factors, and collectively determine the cell type-specific characteristics. Here, we summarize the general properties of TFs, provide an overview of TFs involved in the development and function of the human trophoblast, and address similarities and differences to their murine orthologs. In addition, we discuss how the recent establishment of human in vitro models combined with -omics approaches propel our knowledge and transform the human trophoblast field.
Collapse
|
18
|
Kobayashi EH, Shibata S, Oike A, Kobayashi N, Hamada H, Okae H, Arima T. Genomic imprinting in human placentation. Reprod Med Biol 2022; 21:e12490. [PMID: 36465588 PMCID: PMC9713850 DOI: 10.1002/rmb2.12490] [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: 09/22/2022] [Revised: 10/25/2022] [Accepted: 11/10/2022] [Indexed: 12/02/2022] Open
Abstract
Background Genomic imprinting (GI) is a mammalian-specific epigenetic phenomenon that has been implicated in the evolution of the placenta in mammals. Methods Embryo transfer procedures and trophoblast stem (TS) cells were used to re-examine mouse placenta-specific GI genes. For the analysis of human GI genes, cytotrophoblast cells isolated from human placental tissues were used. Using human TS cells, the biological roles of human GI genes were examined. Main findings (1) Many previously identified mouse GI genes were likely to be falsely identified due to contaminating maternal cells. (2) Human placenta-specific GI genes were comprehensively determined, highlighting incomplete erasure of germline DNA methylation in the human placenta. (3) Human TS cells retained normal GI patterns. (4) Complete hydatidiform mole-derived TS cells were characterized by aberrant GI and enhanced trophoblastic proliferation. The maternally expressed imprinted gene p57KIP2 may be responsible for the enhanced proliferation. (5) The primate-specific microRNA cluster on chromosome 19, which is a placenta-specific GI gene, is essential for self-renewal and differentiation of human TS cells. Conclusion Genomic imprinting plays diverse and important roles in human placentation. Experimental analyses using TS cells suggest that the GI maintenance is necessary for normal placental development in humans.
Collapse
Affiliation(s)
- Eri H. Kobayashi
- Department of Informative GeneticsTohoku University School of MedicineSendaiJapan
| | - Shun Shibata
- Department of Informative GeneticsTohoku University School of MedicineSendaiJapan
| | - Akira Oike
- Department of Informative GeneticsTohoku University School of MedicineSendaiJapan
| | - Norio Kobayashi
- Department of Informative GeneticsTohoku University School of MedicineSendaiJapan
| | - Hirotaka Hamada
- Department of Informative GeneticsTohoku University School of MedicineSendaiJapan
| | - Hiroaki Okae
- Department of Informative GeneticsTohoku University School of MedicineSendaiJapan
| | - Takahiro Arima
- Department of Informative GeneticsTohoku University School of MedicineSendaiJapan
| |
Collapse
|
19
|
Burton GJ, Turco MY. Joan Hunt Senior award lecture: New tools to shed light on the 'black box' of pregnancy. Placenta 2021; 125:54-60. [PMID: 34952691 DOI: 10.1016/j.placenta.2021.12.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/01/2021] [Accepted: 12/16/2021] [Indexed: 01/08/2023]
Abstract
Correct establishment of the placenta is critical to the success of a pregnancy, but many of the key events take place during or shortly after implantation and are inaccessible for study. This inaccessibility, coupled with the lack of a suitable preclinical animal model, means that knowledge of human early placental development and function is extremely limited. Hence, the first trimester is often referred to as the 'black box' of pregnancy. However, recent advances in the derivation of trophoblast stem cells and organoid cultures of the trophoblast and endometrium are opening new opportunities for basic and translational research, providing for the first time cells that faithfully replicate their tissue of origin and proliferate and differentiate in culture in a stable and reproducible manner. These cells are valuable new tools for investigating cell-lineage differentiation and maternal-fetal interactions, but become even more powerful when combined with advances in bioengineering, microfabrication and microfluidic technologies. Assembloids of the endometrium comprising various cell types as model systems to investigate events at implantation, and placentas-on-a-chip for the study of nutrient transfer or drug screening are just two examples. This is a rapidly advancing field that may usher in more personalised approaches to infertility and pregnancy complications. Many of the developments are still at the proof-of-principle phase, but with continued refinement they are likely to shed important light on events that are fundamental to our reproduction as individuals and as a species, yet for ethical reasons are hidden from view.
Collapse
Affiliation(s)
- Graham J Burton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| | - Margherita Y Turco
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| |
Collapse
|
20
|
James JL, Boss AL, Sun C, Allerkamp HH, Clark AR. From stem cells to spiral arteries: A journey through early placental development. Placenta 2021; 125:68-77. [PMID: 34819240 DOI: 10.1016/j.placenta.2021.11.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/11/2021] [Accepted: 11/14/2021] [Indexed: 12/19/2022]
Abstract
Early placental development lays the foundation of a healthy pregnancy, and numerous tightly regulated processes must occur for the placenta to meet the increasing nutrient and oxygen exchange requirements of the growing fetus later in gestation. Inadequacies in early placental development can result in disorders such as fetal growth restriction that do not present clinically until the second half of gestation. Indeed, growth restricted placentae exhibit impaired placental development and function, including reduced overall placental size, decreased branching of villi and the blood vessels within them, altered trophoblast function, and impaired uterine vascular remodelling, which together combine to reduce placental exchange capacity. This review explores the importance of early placental development across multiple anatomical aspects of placentation, from the stem cells and lineage hierarchies from which villous core cells and trophoblasts arise, through extravillous trophoblast invasion and spiral artery remodelling, and finally remodelling of the larger uterine vessels.
Collapse
Affiliation(s)
- Joanna L James
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, New Zealand.
| | - Anna L Boss
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Cherry Sun
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Hanna H Allerkamp
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, New Zealand; Auckland Bioengineering Institute, University of Auckland, New Zealand
| | - Alys R Clark
- Auckland Bioengineering Institute, University of Auckland, New Zealand
| |
Collapse
|
21
|
Perez-Garcia V, Lea G, Lopez-Jimenez P, Okkenhaug H, Burton GJ, Moffett A, Turco MY, Hemberger M. BAP1/ASXL complex modulation regulates epithelial-mesenchymal transition during trophoblast differentiation and invasion. eLife 2021; 10:63254. [PMID: 34170818 PMCID: PMC8233037 DOI: 10.7554/elife.63254] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 06/14/2021] [Indexed: 12/28/2022] Open
Abstract
Normal function of the placenta depends on the earliest developmental stages when trophoblast cells differentiate and invade into the endometrium to establish the definitive maternal-fetal interface. Previously, we identified the ubiquitously expressed tumour suppressor BRCA1-associated protein 1 (BAP1) as a central factor of a novel molecular node controlling early mouse placentation. However, functional insights into how BAP1 regulates trophoblast biology are still missing. Using CRISPR/Cas9 knockout and overexpression technology in mouse trophoblast stem cells, here we demonstrate that the downregulation of BAP1 protein is essential to trigger epithelial-mesenchymal transition (EMT) during trophoblast differentiation associated with a gain of invasiveness. Moreover, we show that the function of BAP1 in suppressing EMT progression is dependent on the binding of BAP1 to additional sex comb-like (ASXL1/2) proteins to form the polycomb repressive deubiquitinase (PR-DUB) complex. Finally, both endogenous expression patterns and BAP1 overexpression experiments in human trophoblast stem cells suggest that the molecular function of BAP1 in regulating trophoblast differentiation and EMT progression is conserved in mice and humans. Our results reveal that the physiological modulation of BAP1 determines the invasive properties of the trophoblast, delineating a new role of the BAP1 PR-DUB complex in regulating early placentation.
Collapse
Affiliation(s)
- Vicente Perez-Garcia
- Epigenetics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom.,Centre for Trophoblast Research, Department of Physiology, Development and Neurosicence, University of Cambridge, Cambridge, United Kingdom.,Centro de Investigación Príncipe Felipe, Eduardo Primo Yúfera, Valencia, Spain.,Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Georgia Lea
- Epigenetics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom
| | | | - Hanneke Okkenhaug
- Epigenetics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom
| | - Graham J Burton
- Centre for Trophoblast Research, Department of Physiology, Development and Neurosicence, University of Cambridge, Cambridge, United Kingdom
| | - Ashley Moffett
- Centre for Trophoblast Research, Department of Physiology, Development and Neurosicence, University of Cambridge, Cambridge, United Kingdom.,Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Margherita Y Turco
- Centre for Trophoblast Research, Department of Physiology, Development and Neurosicence, University of Cambridge, Cambridge, United Kingdom.,Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Myriam Hemberger
- Epigenetics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom.,Centre for Trophoblast Research, Department of Physiology, Development and Neurosicence, University of Cambridge, Cambridge, United Kingdom.,Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada
| |
Collapse
|
22
|
Aplin JD, Jones CJP. Cell dynamics in human villous trophoblast. Hum Reprod Update 2021; 27:904-922. [PMID: 34125187 DOI: 10.1093/humupd/dmab015] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/22/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Villous cytotrophoblast (vCTB) is a precursor cell population that supports the development of syncytiotrophoblast (vSTB), the high surface area barrier epithelium of the placental villus, and the primary interface between maternal and fetal tissue. In light of increasing evidence that the placenta can adapt to changing maternal environments or, under stress, can trigger maternal disease, we consider what properties of these cells empower them to exert a controlling influence on pregnancy progression and outcome. OBJECTIVE AND RATIONALE How are cytotrophoblast proliferation and differentiation regulated in the human placental villus to allow for the increasing demands of the fetal and environmental challenges and stresses that may arise during pregnancy? SEARCH METHODS PubMed was interrogated using relevant keywords and word roots combining trophoblast, villus/villous, syncytio/syncytium, placenta, stem, transcription factor (and the individual genes), signalling, apoptosis, autophagy (and the respective genes) from 1960 to the present. Since removal of trophoblast from its tissue environment is known to fundamentally change cell growth and differentiation kinetics, research that relied exclusively on cell culture has not been the main focus of this review, though it is mentioned where appropriate. Work on non-human placenta is not systematically covered, though mention is made where relevant hypotheses have emerged. OUTCOMES The synthesis of data from the literature has led to a new hypothesis for vCTB dynamics. We propose that a reversible transition can occur from a reserve population in G0 to a mitotically active state. Cells from the in-cycle population can then differentiate irreversibly to intermediate cells that leave the cycle and turn on genes that confer the capacity to fuse with the overlying vSTB as well as other functions associated with syncytial barrier and transport function. We speculate that alterations in the rate of entry to the cell cycle, or return of cells in the mitotic fraction to G0, can occur in response to environmental challenge. We also review evidence on the life cycle of trophoblast from the time that fusion occurs, and point to gaps in knowledge of how large quantities of fetal DNA arrive in maternal circulation. We critique historical methodology and make a case for research to re-address questions about trophoblast lifecycle and dynamics in normal pregnancy and the common diseases of pre-eclampsia and fetal growth restriction, where altered trophoblast kinetics have long been postulated. WIDER IMPLICATIONS The hypothesis requires experimental testing, moving research away from currently accepted methodology towards a new standard that includes representative cell and tissue sampling, assessment of cell cycle and differentiation parameters, and robust classification of cell subpopulations in villous trophoblast, with due attention to gestational age, maternal and fetal phenotype, disease and outcome.
Collapse
Affiliation(s)
- John D Aplin
- Maternal and Fetal Health, University of Manchester, Manchester Academic Health Sciences Centre, St Mary's Hospital, Manchester, UK
| | - Carolyn J P Jones
- Maternal and Fetal Health, University of Manchester, Manchester Academic Health Sciences Centre, St Mary's Hospital, Manchester, UK
| |
Collapse
|
23
|
Lee BK, Kim J. Integrating High-Throughput Approaches and in vitro Human Trophoblast Models to Decipher Mechanisms Underlying Early Human Placenta Development. Front Cell Dev Biol 2021; 9:673065. [PMID: 34150768 PMCID: PMC8206641 DOI: 10.3389/fcell.2021.673065] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/04/2021] [Indexed: 12/13/2022] Open
Abstract
The placenta is a temporary but pivotal organ for human pregnancy. It consists of multiple specialized trophoblast cell types originating from the trophectoderm of the blastocyst stage of the embryo. While impaired trophoblast differentiation results in pregnancy disorders affecting both mother and fetus, the molecular mechanisms underlying early human placenta development have been poorly understood, partially due to the limited access to developing human placentas and the lack of suitable human in vitro trophoblast models. Recent success in establishing human trophoblast stem cells and other human in vitro trophoblast models with their differentiation protocols into more specialized cell types, such as syncytiotrophoblast and extravillous trophoblast, has provided a tremendous opportunity to understand early human placenta development. Unfortunately, while high-throughput research methods and omics tools have addressed numerous molecular-level questions in various research fields, these tools have not been widely applied to the above-mentioned human trophoblast models. This review aims to provide an overview of various omics approaches that can be utilized in the study of human in vitro placenta models by exemplifying some important lessons obtained from omics studies of mouse model systems and introducing recently available human in vitro trophoblast model systems. We also highlight some key unknown questions that might be addressed by such techniques. Integrating high-throughput omics approaches and human in vitro model systems will facilitate our understanding of molecular-level regulatory mechanisms underlying early human placenta development as well as placenta-associated complications.
Collapse
Affiliation(s)
- Bum-Kyu Lee
- Department of Biomedical Sciences, Cancer Research Center, University at Albany-State University of New York, Rensselaer, NY, United States
| | - Jonghwan Kim
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
24
|
Overexpression of miR-210-3p Impairs Extravillous Trophoblast Functions Associated with Uterine Spiral Artery Remodeling. Int J Mol Sci 2021; 22:ijms22083961. [PMID: 33921262 PMCID: PMC8069107 DOI: 10.3390/ijms22083961] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 12/18/2022] Open
Abstract
Hsa-miR-210-3p has been reported to be upregulated in preeclampsia (PE); however, the functions of miR-210-3p in placental development are not fully understood, and, consequently, miR-210-3p’s role in the pathogenesis of PE is still under investigation. In this study, we found that overexpression of miR-210-3p reduced trophoblast migration and invasion, extravillous trophoblast (EVT) outgrowth in first trimester explants, expression of endovascular trophoblast (enEVT) markers and the ability of trophoblast to form endothelial-like networks. In addition, miR-210-3p overexpression significantly downregulated the mRNA levels of interleukin-1B and -8, as well as CXC motif ligand 1. These cytokines have been suggested to play a role in EVT invasion and the recruitment of immune cells to the spiral artery remodeling sites. We also showed that caudal-related homeobox transcription factor 2 (CDX2) is targeted by miR-210-3p and that CDX2 downregulation mimicked the observed effects of miR-210-3p upregulation in trophoblasts. These findings suggest that miR-210-3p may play a role in regulating events associated with enEVT functions and its overexpression could impair spiral artery remodeling, thereby contributing to PE.
Collapse
|
25
|
Two distinct trophectoderm lineage stem cells from human pluripotent stem cells. J Biol Chem 2021; 296:100386. [PMID: 33556374 PMCID: PMC7948510 DOI: 10.1016/j.jbc.2021.100386] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 01/27/2021] [Accepted: 02/03/2021] [Indexed: 01/08/2023] Open
Abstract
The trophectoderm layer of the blastocyst-stage embryo is the precursor for all trophoblast cells in the placenta. Human trophoblast stem (TS) cells have emerged as an attractive tool for studies on early trophoblast development. However, the use of TS cell models is constrained by the limited genetic diversity of existing TS cell lines and restrictions on using human fetal tissue or embryos needed to generate additional lines. Here we report the derivation of two distinct stem cell types of the trophectoderm lineage from human pluripotent stem cells. Analogous to villous cytotrophoblasts in vivo, the first is a CDX2- stem cell comparable with placenta-derived TS cells—they both exhibit identical expression of key markers, are maintained in culture and differentiate under similar conditions, and share high transcriptome similarity. The second is a CDX2+ stem cell with distinct cell culture requirements, and differences in gene expression and differentiation, relative to CDX2- stem cells. Derivation of TS cells from pluripotent stem cells will significantly enable construction of in vitro models for normal and pathological placental development.
Collapse
|
26
|
Burton GJ, Jauniaux E. Placentation in the Human and Higher Primates. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2021; 234:223-254. [PMID: 34694484 DOI: 10.1007/978-3-030-77360-1_11] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Placentation in humans is precocious and highly invasive compared to other mammals. Implantation is interstitial, with the conceptus becoming completely embedded within the endometrium towards the end of the second week post-fertilization. Villi initially form over the entire surface of the chorionic sac, stimulated by histotrophic secretions from the endometrial glands. The secondary yolk sac never makes contact with the chorion, and a choriovitelline placenta is never established. However, recent morphological and transcriptomic analyses suggest that the yolk sac plays an important role in the uptake of nutrients from the coelomic fluid. Measurements performed in vivo demonstrate that early development takes place in a physiological, low-oxygen environment that protects against teratogenic free radicals and maintains stem cells in a multipotent state. The maternal arterial circulation to the placenta is only fully established around 10-12 weeks of gestation. By then, villi have regressed over the superficial, abembryonic pole, leaving the definitive discoid placenta, which is of the villous, hemochorial type. Remodeling of the maternal spiral arteries is essential to ensure a high-volume but low-velocity inflow into the mature placenta. Extravillous trophoblast cells migrate from anchoring villi and surround the arteries. Their interactions with maternal immune cells release cytokines and proteases that are key to remodeling, and a successful pregnancy.
Collapse
Affiliation(s)
- Graham J Burton
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| | - Eric Jauniaux
- Faculty of Population Health Sciences, EGA Institute for Women's Health, University College London, London, UK
| |
Collapse
|
27
|
Io S, Kondoh E, Chigusa Y, Kawasaki K, Mandai M, Yamada AS. New era of trophoblast research: integrating morphological and molecular approaches. Hum Reprod Update 2020; 26:611-633. [PMID: 32728695 DOI: 10.1093/humupd/dmaa020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 03/24/2020] [Accepted: 04/20/2020] [Indexed: 12/27/2022] Open
Abstract
Many pregnancy complications are the result of dysfunction in the placenta. The pathogenic mechanisms of placenta-mediated pregnancy complications, however, are unclear. Abnormal placental development in these conditions begins in the first trimester, but no symptoms are observed during this period. To elucidate effective preventative treatments, understanding the differentiation and development of human placenta is crucial. This review elucidates the uniqueness of the human placenta in early development from the aspect of structural characteristics and molecular markers. We summarise the morphogenesis of human placenta based on human specimens and then compile molecular markers that have been clarified by immunostaining and RNA-sequencing data across species. Relevant studies were identified using the PubMed database and Google Scholar search engines up to March 2020. All articles were independently screened for eligibility by the authors based on titles and abstracts. In particular, the authors carefully examined literature on human placentation. This review integrates the development of human placentation from morphological approaches in comparison with other species and provides new insights into trophoblast molecular markers. The morphological features of human early placentation are described in Carnegie stages (CS), from CS3 (floating blastocyst) to CS9 (emerging point of tertiary villi). Molecular markers are described for each type of trophoblast involved in human placental development. We summarise the character of human trophoblast cell lines and explain how long-term culture system of human cytotrophoblast, both monolayer and spheroid, established in recent studies allows for the generation of human trophoblast cell lines. Due to differences in developmental features among species, it is desirable to understand early placentation in humans. In addition, reliable molecular markers that reflect normal human trophoblast are needed to advance trophoblast research. In the clinical setting, these markers can be valuable means for morphologically and functionally assessing placenta-mediated pregnancy complications and provide early prediction and management of these diseases.
Collapse
Affiliation(s)
- Shingo Io
- Department of Life Science Frontiers, Center for iPS Cell Research & Application, Kyoto University, Kyoto, Japan.,Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Japan Society for the Promotion of Science, Tokyo, Japan
| | - Eiji Kondoh
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yoshitsugu Chigusa
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kaoru Kawasaki
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - And Shigehito Yamada
- Human Health Sciences, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Congenital Anomaly Research Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
28
|
Review: Histotrophic nutrition and the placental-endometrial dialogue during human early pregnancy. Placenta 2020; 102:21-26. [DOI: 10.1016/j.placenta.2020.02.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/07/2020] [Accepted: 02/09/2020] [Indexed: 11/21/2022]
|
29
|
Schmidt JK, Keding LT, Block LN, Wiepz GJ, Koenig MR, Meyer MG, Dusek BM, Kroner KM, Bertogliat MJ, Kallio AR, Mean KD, Golos TG. Placenta-derived macaque trophoblast stem cells: differentiation to syncytiotrophoblasts and extravillous trophoblasts reveals phenotypic reprogramming. Sci Rep 2020; 10:19159. [PMID: 33154556 PMCID: PMC7644694 DOI: 10.1038/s41598-020-76313-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023] Open
Abstract
Nonhuman primates are excellent models for studying human placentation as experimental manipulations in vitro can be translated to in vivo pregnancy. Our objective was to develop macaque trophoblast stem cells (TSCs) as an in vitro platform for future assessment of primate trophoblast development and function. Macaque TSC lines were generated by isolating first and second trimester placental villous cytotrophoblasts followed by culture in TSC medium to maintain cellular proliferation. TSCs grew as mononuclear colonies, whereas upon induction of syncytiotrophoblast (ST) differentiation multinuclear structures appeared, indicative of syncytium formation. Chorionic gonadotropin secretion was > 4000-fold higher in ST culture media compared to TSC media. The secretion of chorionic gonadotropin by TSC-derived ST reflects a reprogramming of macaque TSCs to an earlier pregnancy phenotype. Characteristic trophoblast hallmarks were defined in TSCs and ST including expression of C19MC miRNAs and the macaque placental nonclassical MHC class I molecule, Mamu-AG. Extravillous trophoblasts (EVTs) were derived that express macaque EVT markers Mamu-AG and CD56, and also secrete high levels of MMP2. Our analyses of macaque TSCs suggests that these cells represent a proliferative, self-renewing population capable of differentiating to STs and EVTs in vitro thereby establishing an experimental model of primate placentation.
Collapse
Affiliation(s)
- Jenna Kropp Schmidt
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA.
| | - Logan T Keding
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Lindsey N Block
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Gregory J Wiepz
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Michelle R Koenig
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael G Meyer
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Brittany M Dusek
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Kamryn M Kroner
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Mario J Bertogliat
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Avery R Kallio
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Katherine D Mean
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Thaddeus G Golos
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
30
|
Shibata S, Kobayashi EH, Kobayashi N, Oike A, Okae H, Arima T. Unique features and emerging in vitro models of human placental development. Reprod Med Biol 2020; 19:301-313. [PMID: 33071632 PMCID: PMC7542016 DOI: 10.1002/rmb2.12347] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 08/21/2020] [Accepted: 08/21/2020] [Indexed: 12/13/2022] Open
Abstract
Background The placenta is an essential organ for the normal development of mammalian fetuses. Most of our knowledge on the molecular mechanisms of placental development has come from the analyses of mice, especially histopathological examination of knockout mice. Choriocarcinoma and immortalized cell lines have also been used for basic research on the human placenta. However, these cells are quite different from normal trophoblast cells. Methods In this review, we first provide an overview of mouse and human placental development with particular focus on the differences in the anatomy, transcription factor networks, and epigenetic characteristics between these species. Next, we discuss pregnancy complications associated with abnormal placentation. Finally, we introduce emerging in vitro models to study the human placenta, including human trophoblast stem (TS) cells, trophoblast and endometrium organoids, and artificial embryos. Main findings The placental structure and development differ greatly between humans and mice. The recent establishment of human TS cells and trophoblast and endometrial organoids enhances our understanding of the mechanisms underlying human placental development. Conclusion These in vitro models will greatly advance our understanding of human placental development and potentially contribute to the elucidation of the causes of infertility and other pregnancy complications.
Collapse
Affiliation(s)
- Shun Shibata
- Department of Informative Genetics Tohoku University Graduate School of Medicine Sendai Japan
| | - Eri H Kobayashi
- Department of Informative Genetics Tohoku University Graduate School of Medicine Sendai Japan
| | - Norio Kobayashi
- Department of Informative Genetics Tohoku University Graduate School of Medicine Sendai Japan
| | - Akira Oike
- Department of Informative Genetics Tohoku University Graduate School of Medicine Sendai Japan
| | - Hiroaki Okae
- Department of Informative Genetics Tohoku University Graduate School of Medicine Sendai Japan
| | - Takahiro Arima
- Department of Informative Genetics Tohoku University Graduate School of Medicine Sendai Japan
| |
Collapse
|
31
|
Hoo R, Nakimuli A, Vento-Tormo R. Innate Immune Mechanisms to Protect Against Infection at the Human Decidual-Placental Interface. Front Immunol 2020; 11:2070. [PMID: 33013876 PMCID: PMC7511589 DOI: 10.3389/fimmu.2020.02070] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/29/2020] [Indexed: 12/25/2022] Open
Abstract
During pregnancy, the placenta forms the anatomical barrier between the mother and developing fetus. Infectious agents can potentially breach the placental barrier resulting in pathogenic transmission from mother to fetus. Innate immune responses, orchestrated by maternal and fetal cells at the decidual-placental interface, are the first line of defense to avoid vertical transmission. Here, we outline the anatomy of the human placenta and uterine lining, the decidua, and discuss the potential capacity of pathogen pattern recognition and other host defense strategies present in the innate immune cells at the placental-decidual interface. We consider major congenital infections that access the placenta from hematogenous or decidual route. Finally, we highlight the challenges in studying human placental responses to pathogens and vertical transmission using current experimental models and identify gaps in knowledge that need to be addressed. We further propose novel experimental strategies to address such limitations.
Collapse
Affiliation(s)
- Regina Hoo
- Wellcome Sanger Institute, Cambridge, United Kingdom
- Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| | - Annettee Nakimuli
- Wellcome Sanger Institute, Cambridge, United Kingdom
- Department of Obstetrics and Gynecology, School of Medicine, Makerere University, Kampala, Uganda
| | - Roser Vento-Tormo
- Wellcome Sanger Institute, Cambridge, United Kingdom
- Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
32
|
Saha B, Ganguly A, Home P, Bhattacharya B, Ray S, Ghosh A, Rumi MAK, Marsh C, French VA, Gunewardena S, Paul S. TEAD4 ensures postimplantation development by promoting trophoblast self-renewal: An implication in early human pregnancy loss. Proc Natl Acad Sci U S A 2020; 117:17864-17875. [PMID: 32669432 PMCID: PMC7395512 DOI: 10.1073/pnas.2002449117] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Early pregnancy loss affects ∼15% of all implantation-confirmed human conceptions. However, evolutionarily conserved molecular mechanisms that regulate self-renewal of trophoblast progenitors and their association with early pregnancy loss are poorly understood. Here, we provide evidence that transcription factor TEAD4 ensures survival of postimplantation mouse and human embryos by controlling self-renewal and stemness of trophoblast progenitors within the placenta primordium. In an early postimplantation mouse embryo, TEAD4 is selectively expressed in trophoblast stem cell-like progenitor cells (TSPCs), and loss of Tead4 in postimplantation mouse TSPCs impairs their self-renewal, leading to embryonic lethality before embryonic day 9.0, a developmental stage equivalent to the first trimester of human gestation. Both TEAD4 and its cofactor, yes-associated protein 1 (YAP1), are specifically expressed in cytotrophoblast (CTB) progenitors of a first-trimester human placenta. We also show that a subset of unexplained recurrent pregnancy losses (idiopathic RPLs) is associated with impaired TEAD4 expression in CTB progenitors. Furthermore, by establishing idiopathic RPL patient-specific human trophoblast stem cells (RPL-TSCs), we show that loss of TEAD4 is associated with defective self-renewal in RPL-TSCs and rescue of TEAD4 expression restores their self-renewal ability. Unbiased genomics studies revealed that TEAD4 directly regulates expression of key cell cycle genes in both mouse and human TSCs and establishes a conserved transcriptional program. Our findings show that TEAD4, an effector of the Hippo signaling pathway, is essential for the establishment of pregnancy in a postimplantation mammalian embryo and indicate that impairment of the Hippo signaling pathway could be a molecular cause for early human pregnancy loss.
Collapse
Affiliation(s)
- Biswarup Saha
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160
| | - Avishek Ganguly
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160
| | - Pratik Home
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, KS 66160
| | - Bhaswati Bhattacharya
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160
| | - Soma Ray
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160
| | - Ananya Ghosh
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160
| | - M A Karim Rumi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, KS 66160
| | - Courtney Marsh
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, KS 66160
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Valerie A French
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Sumedha Gunewardena
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Soumen Paul
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160;
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center, Kansas City, KS 66160
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160
| |
Collapse
|
33
|
Ishiuchi T, Ohishi H, Sato T, Kamimura S, Yorino M, Abe S, Suzuki A, Wakayama T, Suyama M, Sasaki H. Zfp281 Shapes the Transcriptome of Trophoblast Stem Cells and Is Essential for Placental Development. Cell Rep 2020; 27:1742-1754.e6. [PMID: 31067460 DOI: 10.1016/j.celrep.2019.04.028] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/13/2019] [Accepted: 04/03/2019] [Indexed: 11/26/2022] Open
Abstract
Placental development is a key event in mammalian reproduction and embryogenesis. However, the molecular basis underlying placental development is not fully understood. Here, we conduct a forward genetic screen to identify regulators for extraembryonic development and identify Zfp281 as a key factor. Zfp281 overexpression in mouse embryonic stem cells facilitates the induction of trophoblast stem-like cells. Zfp281 is preferentially expressed in the undifferentiated trophoblast stem cell population in an FGF-dependent manner, and disruption of Zfp281 in mice causes severe defects in early placental development. Consistently, Zfp281-depleted trophoblast stem cells exhibit defects in maintaining the transcriptome and differentiation capacity. Mechanistically, Zfp281 interacts with MLL or COMPASS subunits and occupies the promoters of its target genes. Importantly, ZNF281, the human ortholog of this factor, is required to stabilize the undifferentiated status of human trophoblast stem cells. Thus, we identify Zfp281 as a conserved factor for the maintenance of trophoblast stem cell plasticity.
Collapse
Affiliation(s)
- Takashi Ishiuchi
- Division of Epigenomics and Development, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan.
| | - Hiroaki Ohishi
- Division of Epigenomics and Development, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Tetsuya Sato
- Division of Bioinformatics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Satoshi Kamimura
- Advanced Biotechnology Center, University of Yamanashi, Yamanashi 400-8510, Japan
| | - Masayoshi Yorino
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Shusaku Abe
- Division of Epigenomics and Development, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Atsushi Suzuki
- Division of Organogenesis and Regeneration, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Teruhiko Wakayama
- Advanced Biotechnology Center, University of Yamanashi, Yamanashi 400-8510, Japan
| | - Mikita Suyama
- Division of Bioinformatics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Hiroyuki Sasaki
- Division of Epigenomics and Development, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan.
| |
Collapse
|
34
|
Horii M, Touma O, Bui T, Parast MM. Modeling human trophoblast, the placental epithelium at the maternal fetal interface. Reproduction 2020; 160:R1-R11. [PMID: 32485667 PMCID: PMC7286067 DOI: 10.1530/rep-19-0428] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 04/17/2020] [Indexed: 12/14/2022]
Abstract
Appropriate human trophoblast lineage specification and differentiation is crucial for the establishment of normal placentation and maintenance of pregnancy. However, due to the lack of proper modeling systems, the molecular mechanisms of these processes are still largely unknown. Much of the early studies in this area have been based on animal models and tumor-derived trophoblast cell lines, both of which are suboptimal for modeling this unique human organ. Recent advances in regenerative and stem cell biology methods have led to development of novel in vitro model systems for studying human trophoblast. These include derivation of human embryonic and induced pluripotent stem cells and establishment of methods for the differentiation of these cells into trophoblast, as well as the more recent derivation of human trophoblast stem cells. In addition, advances in culture conditions, from traditional two-dimensional monolayer culture to 3D culturing systems, have led to development of trophoblast organoid and placenta-on-a-chip model, enabling us to study human trophoblast function in context of more physiologically accurate environment. In this review, we will discuss these various model systems, with a focus on human trophoblast, and their ability to help elucidate the key mechanisms underlying placental development and function. This review focuses on model systems of human trophoblast differentiation, including advantages and limitations of stem cell-based culture, trophoblast organoid, and organ-on-a-chip methods and their applications in understanding placental development and disease.
Collapse
Affiliation(s)
- Mariko Horii
- Department of Pathology, University of California San Diego, La Jolla, California, USA
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California, USA
| | - Ojeni Touma
- Department of Pathology, University of California San Diego, La Jolla, California, USA
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California, USA
| | - Tony Bui
- Department of Pathology, University of California San Diego, La Jolla, California, USA
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California, USA
| | - Mana M Parast
- Department of Pathology, University of California San Diego, La Jolla, California, USA
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
35
|
Dong C, Beltcheva M, Gontarz P, Zhang B, Popli P, Fischer LA, Khan SA, Park KM, Yoon EJ, Xing X, Kommagani R, Wang T, Solnica-Krezel L, Theunissen TW. Derivation of trophoblast stem cells from naïve human pluripotent stem cells. eLife 2020; 9:e52504. [PMID: 32048992 PMCID: PMC7062471 DOI: 10.7554/elife.52504] [Citation(s) in RCA: 184] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 02/11/2020] [Indexed: 12/24/2022] Open
Abstract
Naïve human pluripotent stem cells (hPSCs) provide a unique experimental platform of cell fate decisions during pre-implantation development, but their lineage potential remains incompletely characterized. As naïve hPSCs share transcriptional and epigenomic signatures with trophoblast cells, it has been proposed that the naïve state may have enhanced predisposition for differentiation along this extraembryonic lineage. Here we examined the trophoblast potential of isogenic naïve and primed hPSCs. We found that naïve hPSCs can directly give rise to human trophoblast stem cells (hTSCs) and undergo further differentiation into both extravillous and syncytiotrophoblast. In contrast, primed hPSCs do not support hTSC derivation, but give rise to non-self-renewing cytotrophoblasts in response to BMP4. Global transcriptome and chromatin accessibility analyses indicate that hTSCs derived from naïve hPSCs are similar to blastocyst-derived hTSCs and acquire features of post-implantation trophectoderm. The derivation of hTSCs from naïve hPSCs will enable elucidation of early mechanisms that govern normal human trophoblast development and associated pathologies.
Collapse
Affiliation(s)
- Chen Dong
- Department of Developmental Biology, Washington University School of MedicineSt. LouisUnited States
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
| | - Mariana Beltcheva
- Department of Developmental Biology, Washington University School of MedicineSt. LouisUnited States
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
| | - Paul Gontarz
- Department of Developmental Biology, Washington University School of MedicineSt. LouisUnited States
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
| | - Bo Zhang
- Department of Developmental Biology, Washington University School of MedicineSt. LouisUnited States
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
| | - Pooja Popli
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of MedicineSt. LouisUnited States
| | - Laura A Fischer
- Department of Developmental Biology, Washington University School of MedicineSt. LouisUnited States
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
| | - Shafqat A Khan
- Department of Developmental Biology, Washington University School of MedicineSt. LouisUnited States
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
| | - Kyoung-mi Park
- Department of Developmental Biology, Washington University School of MedicineSt. LouisUnited States
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
| | - Eun-Ja Yoon
- Department of Developmental Biology, Washington University School of MedicineSt. LouisUnited States
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
| | - Xiaoyun Xing
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
- Department of Genetics, Center for Genome Sciences & Systems Biology, Washington University School of MedicineSt. LouisUnited States
| | - Ramakrishna Kommagani
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of MedicineSt. LouisUnited States
| | - Ting Wang
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
- Department of Genetics, Center for Genome Sciences & Systems Biology, Washington University School of MedicineSt. LouisUnited States
| | - Lilianna Solnica-Krezel
- Department of Developmental Biology, Washington University School of MedicineSt. LouisUnited States
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
| | - Thorold W Theunissen
- Department of Developmental Biology, Washington University School of MedicineSt. LouisUnited States
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
| |
Collapse
|
36
|
Szilagyi A, Gelencser Z, Romero R, Xu Y, Kiraly P, Demeter A, Palhalmi J, Gyorffy BA, Juhasz K, Hupuczi P, Kekesi KA, Meinhardt G, Papp Z, Draghici S, Erez O, Tarca AL, Knöfler M, Than NG. Placenta-Specific Genes, Their Regulation During Villous Trophoblast Differentiation and Dysregulation in Preterm Preeclampsia. Int J Mol Sci 2020; 21:ijms21020628. [PMID: 31963593 PMCID: PMC7013556 DOI: 10.3390/ijms21020628] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 12/15/2022] Open
Abstract
The human placenta maintains pregnancy and supports the developing fetus by providing nutrition, gas-waste exchange, hormonal regulation, and an immunological barrier from the maternal immune system. The villous syncytiotrophoblast carries most of these functions and provides the interface between the maternal and fetal circulatory systems. The syncytiotrophoblast is generated by the biochemical and morphological differentiation of underlying cytotrophoblast progenitor cells. The dysfunction of the villous trophoblast development is implicated in placenta-mediated pregnancy complications. Herein, we describe gene modules and clusters involved in the dynamic differentiation of villous cytotrophoblasts into the syncytiotrophoblast. During this process, the immune defense functions are first established, followed by structural and metabolic changes, and then by peptide hormone synthesis. We describe key transcription regulatory molecules that regulate gene modules involved in placental functions. Based on transcriptomic evidence, we infer how villous trophoblast differentiation and functions are dysregulated in preterm preeclampsia, a life-threatening placenta-mediated obstetrical syndrome for the mother and fetus. In the conclusion, we uncover the blueprint for villous trophoblast development and its impairment in preterm preeclampsia, which may aid in the future development of non-invasive biomarkers for placental functions and early identification of women at risk for preterm preeclampsia as well as other placenta-mediated pregnancy complications.
Collapse
Affiliation(s)
- Andras Szilagyi
- Systems Biology of Reproduction Lendulet Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (A.S.); (Z.G.); (P.K.); (A.D.); (J.P.); (K.J.)
| | - Zsolt Gelencser
- Systems Biology of Reproduction Lendulet Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (A.S.); (Z.G.); (P.K.); (A.D.); (J.P.); (K.J.)
| | - Roberto Romero
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD 20692, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (O.E.); (A.L.T.)
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
- Detroit Medical Center, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Florida International University, Miami, FL 33199, USA
| | - Yi Xu
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD 20692, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (O.E.); (A.L.T.)
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Peter Kiraly
- Systems Biology of Reproduction Lendulet Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (A.S.); (Z.G.); (P.K.); (A.D.); (J.P.); (K.J.)
| | - Amanda Demeter
- Systems Biology of Reproduction Lendulet Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (A.S.); (Z.G.); (P.K.); (A.D.); (J.P.); (K.J.)
| | - Janos Palhalmi
- Systems Biology of Reproduction Lendulet Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (A.S.); (Z.G.); (P.K.); (A.D.); (J.P.); (K.J.)
| | - Balazs A. Gyorffy
- Laboratory of Proteomics, Institute of Biology, Eotvos Lorand University, H-1117 Budapest, Hungary; (B.A.G.); (K.A.K.)
| | - Kata Juhasz
- Systems Biology of Reproduction Lendulet Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (A.S.); (Z.G.); (P.K.); (A.D.); (J.P.); (K.J.)
| | - Petronella Hupuczi
- Maternity Private Clinic of Obstetrics and Gynecology, H-1126 Budapest, Hungary; (P.H.); (Z.P.)
| | - Katalin Adrienna Kekesi
- Laboratory of Proteomics, Institute of Biology, Eotvos Lorand University, H-1117 Budapest, Hungary; (B.A.G.); (K.A.K.)
- Department of Physiology and Neurobiology, Eotvos Lorand University, H-1117 Budapest, Hungary
| | - Gudrun Meinhardt
- Department of Obstetrics and Gynecology, Reproductive Biology Unit, Medical University of Vienna, Vienna A-1090, Austria; (G.M.); (M.K.)
| | - Zoltan Papp
- Maternity Private Clinic of Obstetrics and Gynecology, H-1126 Budapest, Hungary; (P.H.); (Z.P.)
- Department of Obstetrics and Gynecology, Semmelweis University, H-1088 Budapest, Hungary
| | - Sorin Draghici
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI 48202, USA;
| | - Offer Erez
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD 20692, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (O.E.); (A.L.T.)
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel
| | - Adi Laurentiu Tarca
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD 20692, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (O.E.); (A.L.T.)
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Martin Knöfler
- Department of Obstetrics and Gynecology, Reproductive Biology Unit, Medical University of Vienna, Vienna A-1090, Austria; (G.M.); (M.K.)
| | - Nandor Gabor Than
- Systems Biology of Reproduction Lendulet Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (A.S.); (Z.G.); (P.K.); (A.D.); (J.P.); (K.J.)
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD 20692, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (O.E.); (A.L.T.)
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Maternity Private Clinic of Obstetrics and Gynecology, H-1126 Budapest, Hungary; (P.H.); (Z.P.)
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary
- Correspondence: ; Tel.: +36-1-382-6788
| |
Collapse
|
37
|
Abstract
The placenta is essential for normal in utero development in mammals. In humans, defective placental formation underpins common pregnancy disorders such as pre-eclampsia and fetal growth restriction. The great variation in placental types across mammals means that animal models have been of limited use in understanding human placental development. However, new tools for studying human placental development, including 3D organoids, stem cell culture systems and single cell RNA sequencing, have brought new insights into this field. Here, we review the morphological, molecular and functional aspects of human placental formation, with a focus on the defining cell of the placenta - the trophoblast.
Collapse
Affiliation(s)
- Margherita Y Turco
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
- Department of Physiology, Neuroscience and Development, University of Cambridge, Cambridge CB2 3EG, UK
| | - Ashley Moffett
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| |
Collapse
|
38
|
Chhabra S, Liu L, Goh R, Kong X, Warmflash A. Dissecting the dynamics of signaling events in the BMP, WNT, and NODAL cascade during self-organized fate patterning in human gastruloids. PLoS Biol 2019; 17:e3000498. [PMID: 31613879 PMCID: PMC6814242 DOI: 10.1371/journal.pbio.3000498] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 10/25/2019] [Accepted: 09/23/2019] [Indexed: 12/22/2022] Open
Abstract
During gastrulation, the pluripotent epiblast self-organizes into the 3 germ layers-endoderm, mesoderm and ectoderm, which eventually form the entire embryo. Decades of research in the mouse embryo have revealed that a signaling cascade involving the Bone Morphogenic Protein (BMP), WNT, and NODAL pathways is necessary for gastrulation. In vivo, WNT and NODAL ligands are expressed near the site of gastrulation in the posterior of the embryo, and knockout of these ligands leads to a failure to gastrulate. These data have led to the prevailing view that a signaling gradient in WNT and NODAL underlies patterning during gastrulation; however, the activities of these pathways in space and time have never been directly observed. In this study, we quantify BMP, WNT, and NODAL signaling dynamics in an in vitro model of human gastrulation. Our data suggest that BMP signaling initiates waves of WNT and NODAL signaling activity that move toward the colony center at a constant rate. Using a simple mathematical model, we show that this wave-like behavior is inconsistent with a reaction-diffusion-based Turing system, indicating that there is no stable signaling gradient of WNT/NODAL. Instead, the final signaling state is homogeneous, and spatial differences arise only from boundary effects. We further show that the durations of WNT and NODAL signaling control mesoderm differentiation, while the duration of BMP signaling controls differentiation of CDX2-positive extra-embryonic cells. The identity of these extra-embryonic cells has been controversial, and we use RNA sequencing (RNA-seq) to obtain their transcriptomes and show that they closely resemble human trophoblast cells in vivo. The domain of BMP signaling is identical to the domain of differentiation of these trophoblast-like cells; however, neither WNT nor NODAL forms a spatial pattern that maps directly to the mesodermal region, suggesting that mesoderm differentiation is controlled dynamically by the combinatorial effect of multiple signals. We synthesize our data into a mathematical model that accurately recapitulates signaling dynamics and predicts cell fate patterning upon chemical and physical perturbations. Taken together, our study shows that the dynamics of signaling events in the BMP, WNT, and NODAL cascade in the absence of a stable signaling gradient control fate patterning of human gastruloids.
Collapse
Affiliation(s)
- Sapna Chhabra
- Systems, Synthetic and Physical Biology, Rice University, Houston, Texas, United States of America
| | - Lizhong Liu
- Department of Biosciences, Rice University, Houston, Texas, United States of America
| | - Ryan Goh
- Department of Mathematics, Boston University, Boston, Massachusetts, United States of America
| | - Xiangyu Kong
- Department of Biosciences, Rice University, Houston, Texas, United States of America
| | - Aryeh Warmflash
- Department of Biosciences, Rice University, Houston, Texas, United States of America
- Department of Bioengineering, Rice University, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
39
|
Frontier Progress in the Establishment of Trophoblast Stem Cell and the Identification of New Cell Subtypes at the Maternal-Fetal Interface. MATERNAL-FETAL MEDICINE 2019. [DOI: 10.1097/fm9.0000000000000023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
40
|
Hemberger M, Hanna CW, Dean W. Mechanisms of early placental development in mouse and humans. Nat Rev Genet 2019; 21:27-43. [PMID: 31534202 DOI: 10.1038/s41576-019-0169-4] [Citation(s) in RCA: 249] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2019] [Indexed: 02/08/2023]
Abstract
The importance of the placenta in supporting mammalian development has long been recognized, but our knowledge of the molecular, genetic and epigenetic requirements that underpin normal placentation has remained remarkably under-appreciated. Both the in vivo mouse model and in vitro-derived murine trophoblast stem cells have been invaluable research tools for gaining insights into these aspects of placental development and function, with recent studies starting to reshape our view of how a unique epigenetic environment contributes to trophoblast differentiation and placenta formation. These advances, together with recent successes in deriving human trophoblast stem cells, open up new and exciting prospects in basic and clinical settings that will help deepen our understanding of placental development and associated disorders of pregnancy.
Collapse
Affiliation(s)
- Myriam Hemberger
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Canada. .,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada. .,Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Canada. .,Epigenetics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, UK. .,Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| | - Courtney W Hanna
- Epigenetics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, UK.,Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Wendy Dean
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, Canada. .,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada. .,Epigenetics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, UK. .,Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Calgary, Canada.
| |
Collapse
|
41
|
Knöfler M, Haider S, Saleh L, Pollheimer J, Gamage TKJB, James J. Human placenta and trophoblast development: key molecular mechanisms and model systems. Cell Mol Life Sci 2019; 76:3479-3496. [PMID: 31049600 PMCID: PMC6697717 DOI: 10.1007/s00018-019-03104-6] [Citation(s) in RCA: 376] [Impact Index Per Article: 75.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/08/2019] [Accepted: 04/11/2019] [Indexed: 12/16/2022]
Abstract
Abnormal placentation is considered as an underlying cause of various pregnancy complications such as miscarriage, preeclampsia and intrauterine growth restriction, the latter increasing the risk for the development of severe disorders in later life such as cardiovascular disease and type 2 diabetes. Despite their importance, the molecular mechanisms governing human placental formation and trophoblast cell lineage specification and differentiation have been poorly unravelled, mostly due to the lack of appropriate cellular model systems. However, over the past few years major progress has been made by establishing self-renewing human trophoblast stem cells and 3-dimensional organoids from human blastocysts and early placental tissues opening the path for detailed molecular investigations. Herein, we summarize the present knowledge about human placental development, its stem cells, progenitors and differentiated cell types in the trophoblast epithelium and the villous core. Anatomy of the early placenta, current model systems, and critical key regulatory factors and signalling cascades governing placentation will be elucidated. In this context, we will discuss the role of the developmental pathways Wingless and Notch, controlling trophoblast stemness/differentiation and formation of invasive trophoblast progenitors, respectively.
Collapse
Affiliation(s)
- Martin Knöfler
- Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, Währinger Gürtel 18-20, 5Q, 1090, Vienna, Austria.
| | - Sandra Haider
- Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, Währinger Gürtel 18-20, 5Q, 1090, Vienna, Austria
| | - Leila Saleh
- Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, Währinger Gürtel 18-20, 5Q, 1090, Vienna, Austria
| | - Jürgen Pollheimer
- Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, Währinger Gürtel 18-20, 5Q, 1090, Vienna, Austria
| | - Teena K J B Gamage
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Joanna James
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
42
|
Soares MJ, Varberg KM, Iqbal K. Hemochorial placentation: development, function, and adaptations. Biol Reprod 2019; 99:196-211. [PMID: 29481584 DOI: 10.1093/biolre/ioy049] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/21/2018] [Indexed: 11/12/2022] Open
Abstract
Placentation is a reproductive adaptation that permits fetal growth and development within the protected confines of the female reproductive tract. Through this important role, the placenta also determines postnatal health and susceptibility to disease. The hemochorial placenta is a prominent feature in primate and rodent development. This manuscript provides an overview of the basics of hemochorial placental development and function, provides perspectives on major discoveries that have shaped placental research, and thoughts on strategies for future investigation.
Collapse
Affiliation(s)
- Michael J Soares
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA.,Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas, USA and the Center for Perinatal Research, Children΄s Research Institute, Children΄s Mercy, Kansas City, Missouri, USA
| | - Kaela M Varberg
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Khursheed Iqbal
- Institute for Reproduction and Perinatal Research and the Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
43
|
High Proliferative Placenta-Derived Multipotent Cells Express Cytokeratin 7 at Low Level. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2098749. [PMID: 31392209 PMCID: PMC6662495 DOI: 10.1155/2019/2098749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 05/30/2019] [Accepted: 06/25/2019] [Indexed: 12/11/2022]
Abstract
The purpose of this study was to investigate the immunophenotypes and gene expression profile of high proliferative placenta-derived multipotent cells (PDMCs) population at different stages of culture. We demonstrated that the colonies resulting from single cells were either positive or negative for CK7, whereas only PDMC clones with weak CK7 expression (CK7low-clones) were highly proliferative. Interestingly, vimentin positive (Vim+) placental stromal mesenchymal cells did not express CK7 in situ, but double CK7+Vim+ cells detection in tissue explants and explants outgrowth indicated CK7 inducible expression in vitro. PCNA presence in CK7+Vim+ cells during placental explants culturing confirmed belonging of these cells to proliferative subpopulation. Transcription factors CDX2 and EOMES were expressed in both CK7low-clones and subset of stromal mesenchymal cells of first-trimester placental tissue in situ. Meanwhile, CK7low -clones and stromal mesenchymal cells of full-term placental tissue in situ expressed ERG heterogeneously. SPP1, COL2A1, and PPARG2 mesodermal-related genes expression by CK7low-clones additionally confirms their mesenchymal origin. Inherent stem cell-related gene expression (IFTM3, POU5F1, and VASA) in CK7low-clones might indicate their enrichment for progenitors. Finally, in CK7low-clones we observed expression of such trophoblast-associated genes as CGB types I and II, fusogenic ERVW-1, GCM1, and GATA3. Thus, our results indicate that PDMCs acquired the representative immunophenotype signature under culture conditions.
Collapse
|
44
|
Gao X, Nowak-Imialek M, Chen X, Chen D, Herrmann D, Ruan D, Chen ACH, Eckersley-Maslin MA, Ahmad S, Lee YL, Kobayashi T, Ryan D, Zhong J, Zhu J, Wu J, Lan G, Petkov S, Yang J, Antunes L, Campos LS, Fu B, Wang S, Yong Y, Wang X, Xue SG, Ge L, Liu Z, Huang Y, Nie T, Li P, Wu D, Pei D, Zhang Y, Lu L, Yang F, Kimber SJ, Reik W, Zou X, Shang Z, Lai L, Surani A, Tam PPL, Ahmed A, Yeung WSB, Teichmann SA, Niemann H, Liu P. Establishment of porcine and human expanded potential stem cells. Nat Cell Biol 2019; 21:687-699. [PMID: 31160711 PMCID: PMC7035105 DOI: 10.1038/s41556-019-0333-2] [Citation(s) in RCA: 236] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 04/24/2019] [Indexed: 12/14/2022]
Abstract
We recently derived mouse expanded potential stem cells (EPSCs) from individual blastomeres by inhibiting the critical molecular pathways that predispose their differentiation. EPSCs had enriched molecular signatures of blastomeres and possessed developmental potency for all embryonic and extra-embryonic cell lineages. Here, we report the derivation of porcine EPSCs, which express key pluripotency genes, are genetically stable, permit genome editing, differentiate to derivatives of the three germ layers in chimeras and produce primordial germ cell-like cells in vitro. Under similar conditions, human embryonic stem cells and induced pluripotent stem cells can be converted, or somatic cells directly reprogrammed, to EPSCs that display the molecular and functional attributes reminiscent of porcine EPSCs. Importantly, trophoblast stem-cell-like cells can be generated from both human and porcine EPSCs. Our pathway-inhibition paradigm thus opens an avenue for generating mammalian pluripotent stem cells, and EPSCs present a unique cellular platform for translational research in biotechnology and regenerative medicine.
Collapse
Affiliation(s)
- Xuefei Gao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Stem Cell and Regenerative Medicine Consortium, Pokfulam, Hong Kong
- The Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Monika Nowak-Imialek
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut (FLI), Mariensee, Neustadt, Germany
- REBIRTH Centre of Excellence, Hannover Medical School, Hannover, Germany
| | - Xi Chen
- The Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Dongsheng Chen
- BGI-Shenzhen, Shenzhen, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Doris Herrmann
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut (FLI), Mariensee, Neustadt, Germany
- REBIRTH Centre of Excellence, Hannover Medical School, Hannover, Germany
| | - Degong Ruan
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Stem Cell and Regenerative Medicine Consortium, Pokfulam, Hong Kong
- Key Laboratory of Regenerative Biology of Chinese Academy of Sciences, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Andy Chun Hang Chen
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | | | - Shakil Ahmad
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham, UK
| | - Yin Lau Lee
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Toshihiro Kobayashi
- Wellcome Trust and Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
| | - David Ryan
- The Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Jixing Zhong
- BGI-Shenzhen, Shenzhen, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Jiacheng Zhu
- BGI-Shenzhen, Shenzhen, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Jian Wu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Stem Cell and Regenerative Medicine Consortium, Pokfulam, Hong Kong
| | - Guocheng Lan
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Stoyan Petkov
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut (FLI), Mariensee, Neustadt, Germany
- REBIRTH Centre of Excellence, Hannover Medical School, Hannover, Germany
- German Primate Center, Platform Degenerative Diseases, Gottingen, Germany
| | - Jian Yang
- The Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liliana Antunes
- The Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Lia S Campos
- The Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Beiyuan Fu
- The Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Shengpeng Wang
- BGI-Shenzhen, Shenzhen, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Yu Yong
- The Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Xiaomin Wang
- Key Laboratory of Regenerative Biology of Chinese Academy of Sciences, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Song-Guo Xue
- Center for Reproductive Medicine, Shanghai East Hospital, School of Medicine, Tong Ji University, Shanghai, China
| | - Liangpeng Ge
- Chongqing Academy of Animal Sciences and Key Laboratory of Pig Industry Sciences, Department of Agriculture, Chongqing, China
| | - Zuohua Liu
- Chongqing Academy of Animal Sciences and Key Laboratory of Pig Industry Sciences, Department of Agriculture, Chongqing, China
| | - Yong Huang
- Chongqing Academy of Animal Sciences and Key Laboratory of Pig Industry Sciences, Department of Agriculture, Chongqing, China
| | - Tao Nie
- Key Laboratory of Regenerative Biology of Chinese Academy of Sciences, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Peng Li
- Key Laboratory of Regenerative Biology of Chinese Academy of Sciences, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Donghai Wu
- Key Laboratory of Regenerative Biology of Chinese Academy of Sciences, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Duanqing Pei
- Key Laboratory of Regenerative Biology of Chinese Academy of Sciences, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Liming Lu
- Institute of Immunology, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Fengtang Yang
- The Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Susan J Kimber
- Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK
| | - Wolf Reik
- Epigenetics Programme, Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Xiangang Zou
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Zhouchun Shang
- BGI-Shenzhen, Shenzhen, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Liangxue Lai
- Key Laboratory of Regenerative Biology of Chinese Academy of Sciences, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Azim Surani
- Wellcome Trust and Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK
| | - Patrick P L Tam
- Embryology Unit, Children's Medical Research Institute and School of Medical Sciences, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Asif Ahmed
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham, UK
| | - William Shu Biu Yeung
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Sarah A Teichmann
- The Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Heiner Niemann
- Institute of Farm Animal Genetics, Friedrich-Loeffler-Institut (FLI), Mariensee, Neustadt, Germany.
- REBIRTH Centre of Excellence, Hannover Medical School, Hannover, Germany.
- Hannover Medical School (MHH), TwinCore, Hannover, Germany.
| | - Pentao Liu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Stem Cell and Regenerative Medicine Consortium, Pokfulam, Hong Kong.
- The Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK.
| |
Collapse
|
45
|
Abstract
Gestational trophoblastic disease or neoplasia covers a spectrum of benign and malignant conditions arising from pregnancies with highly abnormal development of trophoblastic tissue. In this brief review, we discuss the different features of these different conditions and their origins and risk factors and introduce some of the more novel and controversial treatment options currently being explored.
Collapse
Affiliation(s)
- Fen Ning
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Houmei Hou
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Abraham N. Morse
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Gendie E. Lash
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
46
|
Monteiro-Reis S, Lobo J, Henrique R, Jerónimo C. Epigenetic Mechanisms Influencing Epithelial to Mesenchymal Transition in Bladder Cancer. Int J Mol Sci 2019; 20:E297. [PMID: 30642115 PMCID: PMC6358899 DOI: 10.3390/ijms20020297] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/02/2019] [Accepted: 01/09/2019] [Indexed: 12/27/2022] Open
Abstract
Bladder cancer is one of the most incident neoplasms worldwide, and its treatment remains a significant challenge, since the mechanisms underlying disease progression are still poorly understood. The epithelial to mesenchymal transition (EMT) has been proven to play an important role in the tumorigenic process, particularly in cancer cell invasiveness and metastatic potential. Several studies have reported the importance of epigenetic mechanisms and enzymes, which orchestrate them in several features of cancer cells and, specifically, in EMT. In this paper, we discuss the epigenetic enzymes, protein-coding and non-coding genes, and mechanisms altered in the EMT process occurring in bladder cancer cells, as well as its implications, which allows for improved understanding of bladder cancer biology and for the development of novel targeted therapies.
Collapse
Affiliation(s)
- Sara Monteiro-Reis
- Cancer Biology and Epigenetics Group, Research Center, Portuguese Oncology Institute of Porto (CI-IPOP), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.
| | - João Lobo
- Cancer Biology and Epigenetics Group, Research Center, Portuguese Oncology Institute of Porto (CI-IPOP), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.
- Department of Pathology, Portuguese Oncology Institute of Porto (IPOP), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal.
| | - Rui Henrique
- Cancer Biology and Epigenetics Group, Research Center, Portuguese Oncology Institute of Porto (CI-IPOP), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.
- Department of Pathology, Portuguese Oncology Institute of Porto (IPOP), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal.
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, Research Center, Portuguese Oncology Institute of Porto (CI-IPOP), R. Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal.
- Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal.
| |
Collapse
|
47
|
Wong FT, Lin C, Cox BJ. Cellular systems biology identifies dynamic trophoblast populations in early human placentas. Placenta 2019; 76:10-18. [DOI: 10.1016/j.placenta.2018.12.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/03/2018] [Accepted: 12/31/2018] [Indexed: 01/02/2023]
|
48
|
Gamage TKJB, Schierding W, Hurley D, Tsai P, Ludgate JL, Bhoothpur C, Chamley LW, Weeks RJ, Macaulay EC, James JL. The role of DNA methylation in human trophoblast differentiation. Epigenetics 2018; 13:1154-1173. [PMID: 30475094 DOI: 10.1080/15592294.2018.1549462] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The placenta is a vital fetal exchange organ connecting mother and baby. Specialised placental epithelial cells, called trophoblasts, are essential for adequate placental function. Trophoblasts transform the maternal vasculature to allow efficient blood flow to the placenta and facilitate adequate nutrient uptake. Placental development is in part regulated by epigenetic mechanisms. However, our understanding of how DNA methylation contributes to human trophoblast differentiation is limited. To better understand how genome-wide methylation differences affect trophoblast differentiation, reduced representation bisulfite sequencing (RRBS) was conducted on four matched sets of trophoblasts; side-population trophoblasts (a candidate human trophoblast stem cell population), cytotrophoblasts (an intermediate progenitor population), and extravillous trophoblasts (EVT, a terminally differentiated population) each isolated from the same first trimester placenta. Each trophoblast population had a distinct methylome. In line with their close differentiation relationship, the methylation profile of side-population trophoblasts was most similar to cytotrophoblasts, whilst EVT had the most distinct methylome. In comparison to mature trophoblast populations, side-population trophoblasts exhibited differential methylation of genes and miRNAs involved in cell cycle regulation, differentiation, and regulation of pluripotency. A combined methylomic and transcriptomic approach was taken to better understand cytotrophoblast differentiation to EVT. This revealed methylation of 41 genes involved in epithelial to mesenchymal transition and metastatic cancer pathways, which likely contributes to the acquisition of an invasive EVT phenotype. However, the methylation status of a gene did not always predict gene expression. Therefore, while CpG methylation plays a role in trophoblast differentiation, it is likely not the only regulatory mechanism involved in this process.
Collapse
Affiliation(s)
- Teena K J B Gamage
- a Department of Obstetrics and Gynaecology , The University of Auckland , Auckland , New Zealand
| | - William Schierding
- a Department of Obstetrics and Gynaecology , The University of Auckland , Auckland , New Zealand
| | - Daniel Hurley
- b Systems Biology Laboratory, Melbourne School of Engineering , University of Melbourne , Melbourne , Australia
| | - Peter Tsai
- a Department of Obstetrics and Gynaecology , The University of Auckland , Auckland , New Zealand
| | - Jackie L Ludgate
- c Department of Pathology, Dunedin School of Medicine , University of Otago , Dunedin , New Zealand
| | | | - Lawrence W Chamley
- a Department of Obstetrics and Gynaecology , The University of Auckland , Auckland , New Zealand
| | - Robert J Weeks
- c Department of Pathology, Dunedin School of Medicine , University of Otago , Dunedin , New Zealand
| | - Erin C Macaulay
- c Department of Pathology, Dunedin School of Medicine , University of Otago , Dunedin , New Zealand
| | - Joanna L James
- a Department of Obstetrics and Gynaecology , The University of Auckland , Auckland , New Zealand
| |
Collapse
|
49
|
Turco MY, Gardner L, Kay RG, Hamilton RS, Prater M, Hollinshead MS, McWhinnie A, Esposito L, Fernando R, Skelton H, Reimann F, Gribble FM, Sharkey A, Marsh SGE, O'Rahilly S, Hemberger M, Burton GJ, Moffett A. Trophoblast organoids as a model for maternal-fetal interactions during human placentation. Nature 2018; 564:263-267. [PMID: 30487605 PMCID: PMC7220805 DOI: 10.1038/s41586-018-0753-3] [Citation(s) in RCA: 376] [Impact Index Per Article: 62.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 10/31/2018] [Indexed: 11/09/2022]
Abstract
The placenta is the extraembryonic organ that supports the fetus during intrauterine life. Although placental dysfunction results in major disorders of pregnancy with immediate and lifelong consequences for the mother and child, our knowledge of the human placenta is limited owing to a lack of functional experimental models1. After implantation, the trophectoderm of the blastocyst rapidly proliferates and generates the trophoblast, the unique cell type of the placenta. In vivo, proliferative villous cytotrophoblast cells differentiate into two main sub-populations: syncytiotrophoblast, the multinucleated epithelium of the villi responsible for nutrient exchange and hormone production, and extravillous trophoblast cells, which anchor the placenta to the maternal decidua and transform the maternal spiral arteries2. Here we describe the generation of long-term, genetically stable organoid cultures of trophoblast that can differentiate into both syncytiotrophoblast and extravillous trophoblast. We used human leukocyte antigen (HLA) typing to confirm that the organoids were derived from the fetus, and verified their identities against four trophoblast-specific criteria3. The cultures organize into villous-like structures, and we detected the secretion of placental-specific peptides and hormones, including human chorionic gonadotropin (hCG), growth differentiation factor 15 (GDF15) and pregnancy-specific glycoprotein (PSG) by mass spectrometry. The organoids also differentiate into HLA-G+ extravillous trophoblast cells, which vigorously invade in three-dimensional cultures. Analysis of the methylome reveals that the organoids closely resemble normal first trimester placentas. This organoid model will be transformative for studying human placental development and for investigating trophoblast interactions with the local and systemic maternal environment.
Collapse
Affiliation(s)
- Margherita Y Turco
- Department of Pathology, University of Cambridge, Cambridge, UK.
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| | - Lucy Gardner
- Department of Pathology, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Richard G Kay
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Russell S Hamilton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Malwina Prater
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | | | | | - Laura Esposito
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Ridma Fernando
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Helen Skelton
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Frank Reimann
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Fiona M Gribble
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Andrew Sharkey
- Department of Pathology, University of Cambridge, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Steven G E Marsh
- Anthony Nolan Research Institute, Royal Free Hospital, London, UK
- UCL Cancer Institute, Royal Free Campus, London, UK
| | - Stephen O'Rahilly
- Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Myriam Hemberger
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Epigenetics Programme, The Babraham Institute, Cambridge, UK
| | - Graham J Burton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| | - Ashley Moffett
- Department of Pathology, University of Cambridge, Cambridge, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| |
Collapse
|
50
|
Abstract
Establishing the different lineages of the early mammalian embryo takes place over several days and several rounds of cell divisions from the fertilized egg. The resulting blastocyst contains the pluripotent cells of the epiblast, from which embryonic stem cells can be derived, as well as the extraembryonic lineages required for a mammalian embryo to survive in the uterine environment. The dynamics of the cellular and genetic interactions controlling the initiation and maintenance of these lineages in the mouse embryo are increasingly well understood through application of the tools of single-cell genomics, gene editing, and in vivo imaging. Exploring the similarities and differences between mouse and human development will be essential for translation of these findings into new insights into human biology, derivation of stem cells, and improvements in fertility treatments.
Collapse
Affiliation(s)
- Janet Rossant
- Program in Stem Cell and Developmental Biology, Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|