51
|
Zhang S, Xu G, Wu J, Liu X, Fan Y, Chen J, Wallace G, Gu Q. Microphysiological Constructs and Systems: Biofabrication Tactics, Biomimetic Evaluation Approaches, and Biomedical Applications. SMALL METHODS 2024; 8:e2300685. [PMID: 37798902 DOI: 10.1002/smtd.202300685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/23/2023] [Indexed: 10/07/2023]
Abstract
In recent decades, microphysiological constructs and systems (MPCs and MPSs) have undergone significant development, ranging from self-organized organoids to high-throughput organ-on-a-chip platforms. Advances in biomaterials, bioinks, 3D bioprinting, micro/nanofabrication, and sensor technologies have contributed to diverse and innovative biofabrication tactics. MPCs and MPSs, particularly tissue chips relevant to absorption, distribution, metabolism, excretion, and toxicity, have demonstrated potential as precise, efficient, and economical alternatives to animal models for drug discovery and personalized medicine. However, current approaches mainly focus on the in vitro recapitulation of the human anatomical structure and physiological-biochemical indices at a single or a few simple levels. This review highlights the recent remarkable progress in MPC and MPS models and their applications. The challenges that must be addressed to assess the reliability, quantify the techniques, and utilize the fidelity of the models are also discussed.
Collapse
Affiliation(s)
- Shuyu Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, China
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine/Department of Fetal Medicine and Prenatal Diagnosis/BioResource Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Guoshi Xu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 100049, China
| | - Juan Wu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 100049, China
| | - Xiao Liu
- Department of Gastroenterology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yong Fan
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine/Department of Fetal Medicine and Prenatal Diagnosis/BioResource Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Jun Chen
- Intelligent Polymer Research Institute, Australian Institute for Innovative Materials, Innovation Campus, University of Wollongong, North Wollongong, NSW, 2500, Australia
| | - Gordon Wallace
- Intelligent Polymer Research Institute, Australian Institute for Innovative Materials, Innovation Campus, University of Wollongong, North Wollongong, NSW, 2500, Australia
| | - Qi Gu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 100049, China
| |
Collapse
|
52
|
Shimizu T, Oike A, Kobayashi EH, Sekiya A, Kobayashi N, Shibata S, Hamada H, Saito M, Yaegashi N, Suyama M, Arima T, Okae H. CRISPR screening in human trophoblast stem cells reveals both shared and distinct aspects of human and mouse placental development. Proc Natl Acad Sci U S A 2023; 120:e2311372120. [PMID: 38085778 PMCID: PMC10742386 DOI: 10.1073/pnas.2311372120] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 11/09/2023] [Indexed: 12/18/2023] Open
Abstract
The placenta serves as the interface between the mother and fetus, facilitating the exchange of gases and nutrients between their separate blood circulation systems. Trophoblasts in the placenta play a central role in this process. Our current understanding of mammalian trophoblast development relies largely on mouse models. However, given the diversification of mammalian placentas, findings from the mouse placenta cannot be readily extrapolated to other mammalian species, including humans. To fill this knowledge gap, we performed CRISPR knockout screening in human trophoblast stem cells (hTSCs). We targeted genes essential for mouse placental development and identified more than 100 genes as critical regulators in both human hTSCs and mouse placentas. Among them, we further characterized in detail two transcription factors, DLX3 and GCM1, and revealed their essential roles in hTSC differentiation. Moreover, a gene function-based comparison between human and mouse trophoblast subtypes suggests that their relationship may differ significantly from previous assumptions based on tissue localization or cellular function. Notably, our data reveal that hTSCs may not be analogous to mouse TSCs or the extraembryonic ectoderm (ExE) in which in vivo TSCs reside. Instead, hTSCs may be analogous to progenitor cells in the mouse ectoplacental cone and chorion. This finding is consistent with the absence of ExE-like structures during human placental development. Our data not only deepen our understanding of human trophoblast development but also facilitate cross-species comparison of mammalian placentas.
Collapse
Affiliation(s)
- Takanori Shimizu
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai980-8575, Japan
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai980-8575, Japan
| | - Akira Oike
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai980-8575, Japan
- Department of Trophoblast Research, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto860-0811, Japan
| | - Eri H. Kobayashi
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai980-8575, Japan
| | - Asato Sekiya
- Department of Trophoblast Research, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto860-0811, Japan
| | - Norio Kobayashi
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai980-8575, Japan
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI48109
| | - Shun Shibata
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai980-8575, Japan
| | - Hirotaka Hamada
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai980-8575, Japan
| | - Masatoshi Saito
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai980-8575, Japan
| | - Nobuo Yaegashi
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai980-8575, Japan
| | - Mikita Suyama
- Division of Bioinformatics, Medical Institute of Bioregulation, Kyushu University, Fukuoka812-8582, Japan
| | - Takahiro Arima
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai980-8575, Japan
| | - Hiroaki Okae
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai980-8575, Japan
- Department of Trophoblast Research, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto860-0811, Japan
| |
Collapse
|
53
|
Mao Q, Ye Q, Xu Y, Jiang J, Fan Y, Zhuang L, Liu G, Wang T, Zhang Z, Feng T, Kong S, Lu J, Zhang H, Wang H, Lin CP. Murine trophoblast organoids as a model for trophoblast development and CRISPR-Cas9 screening. Dev Cell 2023; 58:2992-3008.e7. [PMID: 38056451 DOI: 10.1016/j.devcel.2023.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/27/2023] [Accepted: 11/10/2023] [Indexed: 12/08/2023]
Abstract
The placenta becomes one of the most diversified organs during placental mammal radiation. The main in vitro model for studying mouse trophoblast development is the 2D differentiation model of trophoblast stem cells, which is highly skewed to certain lineages and thus hampers systematic screens. Here, we established culture conditions for the establishment, maintenance, and differentiation of murine trophoblast organoids. Murine trophoblast organoids under the maintenance condition contain stem cell-like populations, whereas differentiated organoids possess various trophoblasts resembling placental ones in vivo. Ablation of Nubpl or Gcm1 in trophoblast organoids recapitulated their deficiency phenotypes in vivo, suggesting that those organoids are valid in vitro models for trophoblast development. Importantly, we performed an efficient CRISPR-Cas9 screening in mouse trophoblast organoids using a focused sgRNA (single guide RNA) library targeting G protein-coupled receptors. Together, our results establish an organoid model to investigate mouse trophoblast development and a practicable approach to performing forward screening in trophoblast lineages.
Collapse
Affiliation(s)
- Qian Mao
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Qinying Ye
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yiwen Xu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jingwei Jiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yunhao Fan
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Lili Zhuang
- Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| | - Guohui Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Tengfei Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Zhenwu Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Teng Feng
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Shuangbo Kong
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Jinhua Lu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Hui Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Haopeng Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| | - Chao-Po Lin
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
54
|
Lin Q, Cao J, Yu J, Zhu Y, Shen Y, Wang S, Wang Y, Liu Z, Chang Y. YAP-mediated trophoblast dysfunction: the common pathway underlying pregnancy complications. Cell Commun Signal 2023; 21:353. [PMID: 38098027 PMCID: PMC10722737 DOI: 10.1186/s12964-023-01371-2] [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: 07/30/2023] [Accepted: 10/29/2023] [Indexed: 12/17/2023] Open
Abstract
Yes-associated protein (YAP) is a pivotal regulator in cellular proliferation, survival, differentiation, and migration, with significant roles in embryonic development, tissue repair, and tumorigenesis. At the maternal-fetal interface, emerging evidence underscores the importance of precisely regulated YAP activity in ensuring successful pregnancy initiation and progression. However, despite the established association between YAP dysregulation and adverse pregnancy outcomes, insights into the impact of aberrant YAP levels in fetal-derived, particularly trophoblast cells, and the ensuing dysfunction at the maternal-fetal interface remain limited. This review comprehensively examines YAP expression and its regulatory mechanisms in trophoblast cells throughout pregnancy. We emphasize its integral role in placental development and maternal-fetal interactions and delve into the correlations between YAP dysregulation and pregnancy complications. A nuanced understanding of YAP's functions during pregnancy could illuminate intricate molecular mechanisms and pave the way for innovative prevention and treatment strategies for pregnancy complications. Video Abstract.
Collapse
Affiliation(s)
- Qimei Lin
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Nankai University Affiliated Maternity Hospital, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, 300100, China
| | - Jiasong Cao
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Nankai University Affiliated Maternity Hospital, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, 300100, China
| | - Jing Yu
- School of Clinical Medicine, Tianjin Medical University, Tianjin, 300070, China
| | - Yu Zhu
- School of Clinical Medicine, Tianjin Medical University, Tianjin, 300070, China
| | - Yongmei Shen
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Nankai University Affiliated Maternity Hospital, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, 300100, China
| | - Shuqi Wang
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Nankai University Affiliated Maternity Hospital, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, 300100, China
| | - Yixin Wang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Zhen Liu
- Academy of Clinical Medicine, Medical College, Tianjin University, Tianjin, 300072, China
| | - Ying Chang
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Nankai University Affiliated Maternity Hospital, Tianjin Central Hospital of Obstetrics and Gynecology, Tianjin, 300100, China.
- Academy of Clinical Medicine, Medical College, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|
55
|
Brumbaugh J, Aguado BA, Lysaght T, Goldstein LSB. Human fetal tissue is critical for biomedical research. Stem Cell Reports 2023; 18:2300-2312. [PMID: 37977142 PMCID: PMC10724055 DOI: 10.1016/j.stemcr.2023.10.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 11/19/2023] Open
Abstract
Human fetal tissue and cells derived from fetal tissue are crucial for biomedical research. Fetal tissues and cells are used to study both normal development and developmental disorders. They are broadly applied in vaccine development and production. Further, research using cells from fetal tissue is instrumental for studying many infectious diseases, including a broad range of viruses. These widespread applications underscore the value of fetal tissue research and reflect an important point: cells derived from fetal tissues have capabilities that cells from other sources do not. In many cases, increased functionality of cells derived from fetal tissues arises from increased proliferative capacity, ability to survive in culture, and developmental potential that is attenuated in adult tissues. This review highlights important, representative applications of fetal tissue for science and medicine.
Collapse
Affiliation(s)
- Justin Brumbaugh
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA; University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO, USA; Charles C. Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Brian A Aguado
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - Tamra Lysaght
- Centre for Biomedical Ethics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lawrence S B Goldstein
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA; Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
56
|
Suzuki D, Lan KC, Takashima Y. Using human pluripotent stem cells to dissect trophoblast development. Curr Opin Genet Dev 2023; 83:102126. [PMID: 37812907 DOI: 10.1016/j.gde.2023.102126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/12/2023] [Accepted: 09/18/2023] [Indexed: 10/11/2023]
Abstract
In 2021, we showed that naive human pluripotent stem cells (PSCs) can differentiate into trophoblasts via trophectoderm (TE)-like cells. Since TE is a pre-implantation stage of trophoblasts constituting blastocysts, naive human PSCs are an invaluable tool for understanding the entire process of trophoblast development. It has been reported for many years that primed human PSCs can also differentiate into the trophoblast lineage. The in vitro differentiation of naive and primed human PSCs hints at the possibility that human pre- and even post-implantation epiblasts retain the differentiation potential into the trophoblast lineages in vivo. Here, we review the in vitro specification of trophoblasts from human PSCs. Moreover, we discuss the different trophoblast differentiation pathways from naive and primed PSCs.
Collapse
Affiliation(s)
- Daisuke Suzuki
- Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kuan-Chun Lan
- Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yasuhiro Takashima
- Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan.
| |
Collapse
|
57
|
Peng YH, Hsiao SK, Gupta K, Ruland A, Auernhammer GK, Maitz MF, Boye S, Lattner J, Gerri C, Honigmann A, Werner C, Krieg E. Dynamic matrices with DNA-encoded viscoelasticity for cell and organoid culture. NATURE NANOTECHNOLOGY 2023; 18:1463-1473. [PMID: 37550574 PMCID: PMC10716043 DOI: 10.1038/s41565-023-01483-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 07/10/2023] [Indexed: 08/09/2023]
Abstract
Three-dimensional cell and organoid cultures rely on the mechanical support of viscoelastic matrices. However, commonly used matrix materials lack control over key cell-instructive properties. Here we report on fully synthetic hydrogels based on DNA libraries that self-assemble with ultrahigh-molecular-weight polymers, forming a dynamic DNA-crosslinked matrix (DyNAtrix). DyNAtrix enables computationally predictable and systematic control over its viscoelasticity, thermodynamic and kinetic parameters by changing DNA sequence information. Adjustable heat activation allows homogeneous embedding of mammalian cells. Intriguingly, stress-relaxation times can be tuned over four orders of magnitude, recapitulating mechanical characteristics of living tissues. DyNAtrix is self-healing, printable, exhibits high stability, cyto- and haemocompatibility, and controllable degradation. DyNAtrix-based cultures of human mesenchymal stromal cells, pluripotent stem cells, canine kidney cysts and human trophoblast organoids show high viability, proliferation and morphogenesis. DyNAtrix thus represents a programmable and versatile precision matrix for advanced approaches to biomechanics, biophysics and tissue engineering.
Collapse
Affiliation(s)
- Yu-Hsuan Peng
- Institute for Biofunctional Polymer Materials, Leibniz Institute of Polymer Research Dresden, Dresden, Germany
- Center for Regenerative Therapies Dresden, Cluster of Excellence Physics of Life and Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Syuan-Ku Hsiao
- Institute for Biofunctional Polymer Materials, Leibniz Institute of Polymer Research Dresden, Dresden, Germany
- Center for Regenerative Therapies Dresden, Cluster of Excellence Physics of Life and Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Krishna Gupta
- Institute for Biofunctional Polymer Materials, Leibniz Institute of Polymer Research Dresden, Dresden, Germany
- Center for Regenerative Therapies Dresden, Cluster of Excellence Physics of Life and Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Dresden, Germany
| | - André Ruland
- Institute for Biofunctional Polymer Materials, Leibniz Institute of Polymer Research Dresden, Dresden, Germany
| | - Günter K Auernhammer
- Institute for Physical Chemistry and Polymer Physics, Polymer Interfaces, Leibniz Institute of Polymer Research Dresden, Dresden, Germany
| | - Manfred F Maitz
- Institute for Biofunctional Polymer Materials, Leibniz Institute of Polymer Research Dresden, Dresden, Germany
| | - Susanne Boye
- Institute for Macromolecular Chemistry, Leibniz Institute of Polymer Research Dresden, Dresden, Germany
| | - Johanna Lattner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Center for Systems Biology Dresden, Dresden, Germany
| | - Claudia Gerri
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Center for Systems Biology Dresden, Dresden, Germany
| | - Alf Honigmann
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Center for Systems Biology Dresden, Dresden, Germany
| | - Carsten Werner
- Institute for Biofunctional Polymer Materials, Leibniz Institute of Polymer Research Dresden, Dresden, Germany
- Center for Regenerative Therapies Dresden, Cluster of Excellence Physics of Life and Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Elisha Krieg
- Institute for Biofunctional Polymer Materials, Leibniz Institute of Polymer Research Dresden, Dresden, Germany.
- Center for Regenerative Therapies Dresden, Cluster of Excellence Physics of Life and Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
58
|
Dietrich B, Kunihs V, Lackner AI, Meinhardt G, Koo BK, Pollheimer J, Haider S, Knöfler M. NOTCH3 signalling controls human trophoblast stem cell expansion and differentiation. Development 2023; 150:dev202152. [PMID: 37905445 DOI: 10.1242/dev.202152] [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: 07/03/2023] [Accepted: 10/19/2023] [Indexed: 11/02/2023]
Abstract
Failures in growth and differentiation of the early human placenta are associated with severe pregnancy disorders such as pre-eclampsia and fetal growth restriction. However, regulatory mechanisms controlling development of placental epithelial cells, the trophoblasts, remain poorly elucidated. Using trophoblast stem cells (TSCs), trophoblast organoids (TB-ORGs) and primary cytotrophoblasts (CTBs) of early pregnancy, we herein show that autocrine NOTCH3 signalling controls human placental expansion and differentiation. The NOTCH3 receptor was specifically expressed in proliferative CTB progenitors and its active form, the nuclear NOTCH3 intracellular domain (NOTCH3-ICD), interacted with the transcriptional co-activator mastermind-like 1 (MAML1). Doxycycline-inducible expression of dominant-negative MAML1 in TSC lines provoked cell fusion and upregulation of genes specific for multinucleated syncytiotrophoblasts, which are the differentiated hormone-producing cells of the placenta. However, progenitor expansion and markers of trophoblast stemness and proliferation were suppressed. Accordingly, inhibition of NOTCH3 signalling diminished growth of TB-ORGs, whereas overexpression of NOTCH3-ICD in primary CTBs and TSCs showed opposite effects. In conclusion, the data suggest that canonical NOTCH3 signalling plays a key role in human placental development by promoting self-renewal of CTB progenitors.
Collapse
Affiliation(s)
- Bianca Dietrich
- Placental Development Group, Medical University of Vienna, A-1090 Vienna, Austria
| | - Victoria Kunihs
- Placental Development Group, Medical University of Vienna, A-1090 Vienna, Austria
| | - Andreas I Lackner
- Maternal-Fetal Immunology Group, Department of Obstetrics and Gynecology, Reproductive Biology Unit, Medical University of Vienna, A-1090 Vienna, Austria
| | - Gudrun Meinhardt
- Placental Development Group, Medical University of Vienna, A-1090 Vienna, Austria
| | - Bon-Kyoung Koo
- Center for Genome Engineering, Institute for Basic Science, Yuseong-Gu, Daejeon 34126, Republic of Korea
| | - Jürgen Pollheimer
- Maternal-Fetal Immunology Group, Department of Obstetrics and Gynecology, Reproductive Biology Unit, Medical University of Vienna, A-1090 Vienna, Austria
| | - Sandra Haider
- Placental Development Group, Medical University of Vienna, A-1090 Vienna, Austria
| | - Martin Knöfler
- Placental Development Group, Medical University of Vienna, A-1090 Vienna, Austria
| |
Collapse
|
59
|
Tekkatte C, Lindsay SA, Duggan E, Castro-Martínez A, Hakim A, Saldana I, Zhang Y, Zhou J, Sebastian R, Liu Y, Pontigon DS, Meads M, Liu TN, Pizzo DP, Nolan J, Parast MM, Laurent LC. Identification of optimal conditions for human placental explant culture and extracellular vesicle release. iScience 2023; 26:108046. [PMID: 37829201 PMCID: PMC10565782 DOI: 10.1016/j.isci.2023.108046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 05/17/2023] [Accepted: 09/21/2023] [Indexed: 10/14/2023] Open
Abstract
Extracellular vesicles (EVs) can mediate intercellular communication, including signaling between the placenta and maternal tissues. Human placental explant culture is a versatile in vitro model system to investigate placental function. We performed systematic studies in different tissue culture media types and oxygen tensions to identify a defined serum-free culture condition that supports high trophoblast viability and metabolism, as well as the release of similar populations of EVs, compared to traditional undefined conditions that contain media additives potentially contaminated with exogenous EVs. We also determined the time frame in which trophoblast viability and functionality remain optimal. Multiplex vesicle flow cytometry with classical EV and placenta-specific markers revealed three separate populations of explant-derived EVs: small CD63+ EVs; large PLAP+ EVs; and CD63-/PLAP- EVs. These culture and analytical approaches will enable in vitro modeling of short-term effects of environmental perturbations associated with pregnancy complications on placental function and EV release.
Collapse
Affiliation(s)
- Chandana Tekkatte
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92037, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Scott A. Lindsay
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92037, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Erika Duggan
- Scintillon Institute, San Diego, CA 92121, USA
- Cellarcus Biosciences Inc, La Jolla, CA 92037, USA
| | - Anelizze Castro-Martínez
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92037, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Abbas Hakim
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92037, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Isabella Saldana
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92037, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Yan Zhang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92037, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
- Department of Obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jun Zhou
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92037, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
- Department of Obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Rachel Sebastian
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92037, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Yukun Liu
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92037, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
- Department of Obstetrics and Gynecology, SUN Yat-sen Memorial Hospital, SUN Yat-sen University, Guangzhou, Guangdong, China
| | - Devin S. Pontigon
- Department of Pathology, University of California San Diego, La Jolla, CA 92037, USA
| | - Morgan Meads
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
- Department of Pathology, University of California San Diego, La Jolla, CA 92037, USA
| | - Tzu Ning Liu
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92037, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Donald P. Pizzo
- Department of Pathology, University of California San Diego, La Jolla, CA 92037, USA
| | - John Nolan
- Scintillon Institute, San Diego, CA 92121, USA
- Cellarcus Biosciences Inc, La Jolla, CA 92037, USA
| | - Mana M. Parast
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
- Department of Pathology, University of California San Diego, La Jolla, CA 92037, USA
| | - Louise C. Laurent
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92037, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| |
Collapse
|
60
|
Zhuang BM, Cao DD, Li TX, Liu XF, Lyu MM, Wang SD, Cui XY, Wang L, Chen XL, Lin XL, Lee CL, Chiu PCN, Yeung WSB, Yao YQ. Single-cell characterization of self-renewing primary trophoblast organoids as modeling of EVT differentiation and interactions with decidual natural killer cells. BMC Genomics 2023; 24:618. [PMID: 37853336 PMCID: PMC10583354 DOI: 10.1186/s12864-023-09690-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/20/2023] [Indexed: 10/20/2023] Open
Abstract
BACKGROUND Extravillous trophoblast cell (EVT) differentiation and its communication with maternal decidua especially the leading immune cell type natural killer (NK) cell are critical events for placentation. However, appropriate in vitro modelling system and regulatory programs of these two events are still lacking. Recent trophoblast organoid (TO) has advanced the molecular and mechanistic research in placentation. Here, we firstly generated the self-renewing TO from human placental villous and differentiated it into EVTs (EVT-TO) for investigating the differentiation events. We then co-cultured EVT-TO with freshly isolated decidual NKs for further study of cell communication. TO modelling of EVT differentiation as well as EVT interaction with dNK might cast new aspect for placentation research. RESULTS Single-cell RNA sequencing (scRNA-seq) was applied for comprehensive characterization and molecular exploration of TOs modelling of EVT differentiation and interaction with dNKs. Multiple distinct trophoblast states and dNK subpopulations were identified, representing CTB, STB, EVT, dNK1/2/3 and dNKp. Lineage trajectory and Seurat mapping analysis identified the close resemblance of TO and EVT-TO with the human placenta characteristic. Transcription factors regulatory network analysis revealed the cell-type specific essential TFs for controlling EVT differentiation. CellphoneDB analysis predicted the ligand-receptor complexes in dNK-EVT-TO co-cultures, which relate to cytokines, immunomodulation and angiogenesis. EVT was known to affect the immune properties of dNK. Our study found out that on the other way around, dNKs could exert effects on EVT causing expression changes which are functionally important. CONCLUSION Our study documented a single-cell atlas for TO and its applications on EVT differentiation and communications with dNKs, and thus provide methodology and novel research cues for future study of human placentation.
Collapse
Affiliation(s)
- Bai-Mei Zhuang
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, Guangdong, P.R. China
- Medical school of Chinese People's Liberation Army, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Dan-Dan Cao
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, Guangdong, P.R. China.
| | - Tian-Xi Li
- Geneplus-Shenzhen Institute, Shenzhen, China
| | - Xiao-Feng Liu
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, Guangdong, P.R. China
| | - Min-Min Lyu
- Department of Clinical-Translational and Basic Research Laboratory, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Shenzhen, Futian District, Guangdong, P.R. China
| | - Si-Dong Wang
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, Guangdong, P.R. China
- Medical school of Chinese People's Liberation Army, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xin-Yuan Cui
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, Guangdong, P.R. China
| | - Li Wang
- Department of Obstetrics and Gynecology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Xiao-Lin Chen
- Department of Obstetrics and Gynecology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Xiao-Li Lin
- Department of Obstetrics and Gynecology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Cheuk-Lun Lee
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, Guangdong, P.R. China
- Department of Obstetrics and Gynecology, LKS Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong S.A.R
| | - Philip C N Chiu
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, Guangdong, P.R. China
- Department of Obstetrics and Gynecology, LKS Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong S.A.R
| | - William S B Yeung
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, Guangdong, P.R. China
- Department of Obstetrics and Gynecology, LKS Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong S.A.R
| | - Yuan-Qing Yao
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, Guangdong, P.R. China.
- Medical school of Chinese People's Liberation Army, Chinese People's Liberation Army General Hospital, Beijing, China.
- Department of Obstetrics and Gynecology, The First Medical Centre, Chinese People's Liberation Army General Hospital, Beijing, China.
| |
Collapse
|
61
|
Li X, Li ZH, Wang YX, Liu TH. A comprehensive review of human trophoblast fusion models: recent developments and challenges. Cell Death Discov 2023; 9:372. [PMID: 37816723 PMCID: PMC10564767 DOI: 10.1038/s41420-023-01670-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/23/2023] [Accepted: 09/29/2023] [Indexed: 10/12/2023] Open
Abstract
As an essential component of the maternal-fetal interface, the placental syncytiotrophoblast layer contributes to a successful pregnancy by secreting hormones necessary for pregnancy, transporting nutrients, mediating gas exchange, balancing immune tolerance, and resisting pathogen infection. Notably, the deficiency in mononuclear trophoblast cells fusing into multinucleated syncytiotrophoblast has been linked to adverse pregnancy outcomes, such as preeclampsia, fetal growth restriction, preterm birth, and stillbirth. Despite the availability of many models for the study of trophoblast fusion, there exists a notable disparity from the ideal model, limiting the deeper exploration into the placental development. Here, we reviewed the existing models employed for the investigation of human trophoblast fusion from several aspects, including the development history, latest progress, advantages, disadvantages, scope of application, and challenges. The literature searched covers the monolayer cell lines, primary human trophoblast, placental explants, human trophoblast stem cells, human pluripotent stem cells, three-dimensional cell spheres, organoids, and placenta-on-a-chip from 1938 to 2023. These diverse models have significantly enhanced our comprehension of placental development regulation and the underlying mechanisms of placental-related disorders. Through this review, our objective is to provide readers with a thorough understanding of the existing trophoblast fusion models, making it easier to select most suitable models to address specific experimental requirements or scientific inquiries. Establishment and application of the existing human placental trophoblast fusion models.
Collapse
Affiliation(s)
- Xia Li
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China
- The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, 400016, Chongqing, China
| | - Zhuo-Hang Li
- The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, 400016, Chongqing, China
- Medical Laboratory Department, Traditional Chinese Medicine Hospital of Yaan, 625099, Sichuan, China
| | - Ying-Xiong Wang
- The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, 400016, Chongqing, China.
| | - Tai-Hang Liu
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China.
- The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, 400016, Chongqing, China.
| |
Collapse
|
62
|
Zhou J, Sheridan MA, Tian Y, Dahlgren KJ, Messler M, Peng T, Ezashi T, Schulz LC, Ulery BD, Roberts RM, Schust DJ. Development of properly-polarized trophoblast stem cell-derived organoids to model early human pregnancy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.30.560327. [PMID: 37873440 PMCID: PMC10592868 DOI: 10.1101/2023.09.30.560327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
The development of human trophoblast stem cells (hTSC) and stem cell-derived trophoblast organoids has enabled investigation of placental physiology and disease and early maternal-fetal interactions during a stage of human pregnancy that previously had been severely restricted. A key shortcoming in existing trophoblast organoid methodologies is the non-physiologic position of the syncytiotrophoblast (STB) within the inner portion of the organoid, which neither recapitulates placental villous morphology in vivo nor allows for facile modeling of STB exposure to the endometrium or the contents of the intervillous space. Here we have successfully established properly-polarized human trophoblast stem cell (hTSC)-sourced organoids with STB forming on the surface of the organoid. These organoids can also be induced to give rise to the extravillous trophoblast (EVT) lineage with HLA-G + migratory cells that invade into an extracellular matrix-based hydrogel. Compared to previous hTSC organoid methods, organoids created by this method more closely mimic the architecture of the developing human placenta and provide a novel platform to study normal and abnormal human placental development and to model exposures to pharmaceuticals, pathogens and environmental insults. Motivation Human placental organoids have been generated to mimic physiological cell-cell interactions. However, those published models derived from human trophoblast stem cells (hTSCs) or placental villi display a non-physiologic "inside-out" morphology. In vivo , the placental villi have an outer layer of syncytialized cells that are in direct contact with maternal blood, acting as a conduit for gas and nutrient exchange, and an inner layer of progenitor, single cytotrophoblast cells that fuse to create the syncytiotrophoblast layer. Existing "inside-out" models put the cytotrophoblast cells in contact with culture media and substrate, making physiologic interactions between syncytiotrophoblast and other cells/tissues and normal and pathogenic exposures coming from maternal blood difficult to model. The goal of this study was to develop an hTSC-derived 3-D human trophoblast organoid model that positions the syncytiotrophoblast layer on the outside of the multicellular organoid. Graphical abstract
Collapse
|
63
|
Morey R, Bui T, Fisch KM, Horii M. Modeling placental development and disease using human pluripotent stem cells. Placenta 2023; 141:18-25. [PMID: 36333266 PMCID: PMC10148925 DOI: 10.1016/j.placenta.2022.10.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/03/2022] [Accepted: 10/18/2022] [Indexed: 11/08/2022]
Abstract
Our current knowledge of the cellular and molecular mechanisms of placental epithelial cells, trophoblast, primarily came from the use of mouse trophoblast stem cells and tumor-derived or immortalized human trophoblast cell lines. This was mainly due to the difficulties in maintaining primary trophoblast in culture and establishing human trophoblast stem cell (hTSC) lines. However, in-depth characterization of these cellular models and in vivo human trophoblast have revealed significant discrepancies. For the past two decades, multiple groups have shown that human pluripotent stem cells (hPSCs) can be differentiated into trophoblast, and thus could be used as a model for normal and disease trophoblast differentiation. During this time, trophoblast differentiation protocols have evolved, enabling researchers to study cellular characteristics at trophectoderm (TE), trophoblast stem cells (TSC), syncytiotrophoblast (STB), and extravillous trophoblast (EVT) stages. Recently, several groups reported methods to derive hTSC from pre-implantation blastocyst or early gestation placenta, and trophoblast organoids from early gestation placenta, drastically changing the landscape of trophoblast research. These culture conditions have been rapidly applied to generate hPSC-derived TSC and trophoblast organoids. As a result of these technological advancements, the field's capacity to better understand trophoblast differentiation and their involvement in pregnancy related disease has greatly expanded. Here, we present in vitro models of human trophoblast differentiation, describing both primary and hPSC-derived TSC, maintained as monolayers and 3-dimensional trophoblast organoids, as a tool to study early placental development and disease in multiple settings.
Collapse
Affiliation(s)
- Robert Morey
- 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
| | - 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
| | - Kathleen M Fisch
- Department of Obstetrics, Gynecology, and Reproductive Sciences, 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.
| |
Collapse
|
64
|
Wu H, Huang XY, Sun MX, Wang Y, Zhou HY, Tian Y, He B, Li K, Li DY, Wu AP, Wang H, Qin CF. Zika virus targets human trophoblast stem cells and prevents syncytialization in placental trophoblast organoids. Nat Commun 2023; 14:5541. [PMID: 37684223 PMCID: PMC10491779 DOI: 10.1038/s41467-023-41158-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Zika virus (ZIKV) infection during pregnancy threatens pregnancy and fetal health. However, the infectivity and pathological effects of ZIKV on placental trophoblast progenitor cells in early human embryos remain largely unknown. Here, using human trophoblast stem cells (hTSCs), we demonstrated that hTSCs were permissive to ZIKV infection, and resistance to ZIKV increased with hTSC differentiation. Combining gene knockout and transcriptome analysis, we demonstrated that the intrinsic expression of AXL and TIM-1, and the absence of potent interferon (IFN)-stimulated genes (ISGs) and IFNs contributed to the high sensitivity of hTSCs to ZIKV. Furthermore, using our newly developed hTSC-derived trophoblast organoid (hTSC-organoid), we demonstrated that ZIKV infection disrupted the structure of mature hTSC-organoids and inhibited syncytialization. Single-cell RNA sequencing (scRNA-seq) further demonstrated that ZIKV infection of hTSC-organoids disrupted the stemness of hTSCs and the proliferation of cytotrophoblast cells (CTBs) and probably led to a preeclampsia (PE) phenotype. Overall, our results clearly demonstrate that hTSCs represent the major target cells of ZIKV, and a reduced syncytialization may result from ZIKV infection of early developing placenta. These findings deepen our understanding of the characteristics and consequences of ZIKV infection of hTSCs in early human embryos.
Collapse
Affiliation(s)
- Hao Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xing-Yao Huang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Meng-Xu Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Yue Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hang-Yu Zhou
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, China
| | - Ying Tian
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Beijia He
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kai Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - De-Yu Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Ai-Ping Wu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, 215123, China
| | - Hongmei Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, 100071, China.
- Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing, 100071, China.
| |
Collapse
|
65
|
Huang L, Tu Z, Wei L, Sun W, Wang Y, Bi S, He F, Du L, Chen J, Kzhyshkowska J, Wang H, Chen D, Zhang S. Generating Functional Multicellular Organoids from Human Placenta Villi. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301565. [PMID: 37438660 PMCID: PMC10502861 DOI: 10.1002/advs.202301565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/26/2023] [Indexed: 07/14/2023]
Abstract
The interaction between trophoblasts, stroma cells, and immune cells at the maternal-fetal interface constitutes the functional units of the placenta, which is crucial for successful pregnancy outcomes. However, the investigation of this intricate interplay is restricted due to the absence of efficient experimental models. To address this challenge, a robust, reliable methodology for generating placenta villi organoids (PVOs) from early, late, or diseased pregnancies using air-liquid surface culture is developed. PVOs contain cytotrophoblasts that can self-renew and differentiate directly, along with stromal elements that retain native immune cells. Analysis of scRNA sequencing and WES data reveals that PVOs faithfully recapitulate the cellular components and genetic alterations of the corresponding source tissue. Additionally, PVOs derived from patients with preeclampsia exhibit specific pathological features such as inflammation, antiangiogenic imbalance, and decreased syncytin expression. The PVO-based propagation of primary placenta villi should enable a deeper investigation of placenta development and exploration of the underlying pathogenesis and therapeutics of placenta-originated diseases.
Collapse
Affiliation(s)
- Lijun Huang
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Zhaowei Tu
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Liudan Wei
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Wei Sun
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Yifan Wang
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Shilei Bi
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Fang He
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Lili Du
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Jingsi Chen
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Julia Kzhyshkowska
- Institute of Transfusion Medicine and ImmunologyMedical Faculty MannheimUniversity of Heidelberg68167MannheimGermany
| | - Haibin Wang
- Fujian Provincial Key Laboratory of Reproductive Health ResearchDepartment of Obstetrics and GynecologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamen361005China
| | - Dunjin Chen
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
- Key Laboratory for Major Obstetric Diseases of Guangdong ProvinceGuangzhou510150China
- Guangdong‐Hong Kong‐Macao Greater Bay Area Higher Education Joint Laboratory of Maternal‐Fetal MedicineGuangzhou510150China
- Guangdong Engineering and Technology Research Center of Maternal‐Fetal MedicineGuangzhou510150China
| | - Shuang Zhang
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| |
Collapse
|
66
|
Park JY, Lim H, Qin J, Lee LP. Creating mini-pregnancy models in vitro with clinical perspectives. EBioMedicine 2023; 95:104780. [PMID: 37657136 PMCID: PMC10480532 DOI: 10.1016/j.ebiom.2023.104780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/16/2023] [Accepted: 08/18/2023] [Indexed: 09/03/2023] Open
Abstract
During the last decade, organs-on-chips or organoids microphysiological analysis platforms (MAP) have garnered attention in the practical applications of disease models, drug discovery, and developmental biology. Research on pregnant women has firm limitations due to ethical issues; thus, remodelling human pregnancy in vitro is highly beneficial for treatment modality development via disease remodelling or drug monitoring. This review highlights current efforts in bioengineering devices to reproduce human pregnancy and emphasises the significant convergence of biology, engineering, and maternal-foetal medicine. First, we review recent achievements in culturing cells from tissues involved in pregnancy; specifically, trophoblasts from the placenta. Second, we highlight developments in the reconstitution of pregnancy-related female reproductive organs across several structural and functional interpretations. Last, we examine research on the fundamental comprehension of pregnancy-associated diseases to find bioengineering solutions. Recreating human pregnancy through an engineered model is naturally complex; nevertheless, challenges are inevitable to progress precision medicine.
Collapse
Affiliation(s)
- Jee Yoon Park
- Department of Obstetrics and Gynecology, Seoul National University, Bundang Hospital, Seoul National University College of Medicine, Republic of Korea; Department of Medicine, Harvard Medical School, Brigham Women's Hospital, Boston, MA, USA.
| | - Hosub Lim
- Department of Medicine, Harvard Medical School, Brigham Women's Hospital, Boston, MA, USA
| | - Jianhua Qin
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Luke P Lee
- Department of Medicine, Harvard Medical School, Brigham Women's Hospital, Boston, MA, USA; Department of Bioengineering, Department of Electrical Engineering and Computer Science, University of California at Berkeley, Berkeley, CA, USA; Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Korea.
| |
Collapse
|
67
|
Naama M, Buganim Y. Human trophoblast stem cell-state acquisition from pluripotent stem cells and somatic cells. Curr Opin Genet Dev 2023; 81:102084. [PMID: 37451165 DOI: 10.1016/j.gde.2023.102084] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 06/10/2023] [Accepted: 06/11/2023] [Indexed: 07/18/2023]
Abstract
For an extended period of time, research on human embryo implantation and early placentation was hindered by ethical limitation and lack of appropriate in vitro models. Recently, an explosion of new research has significantly expanded our knowledge of early human trophoblast development and facilitated the derivation and culture of self-renewing human trophoblast stem cells (hTSCs). Multiple approaches have been undertaken in efforts to derive and understand hTSCs, including from blastocysts, early trophoblast tissue, and, more recently, from human pluripotent stem cells (hPSCs) and somatic cells. In this concise review, we summarize recent advances in derivation of hTSCs, with a focus on derivation from naive and primed hPSCs, as well as via reprogramming of somatic cells into induced hTSCs. Each of these methods harbors distinct advantages and setbacks, which are discussed. Finally, we briefly explore the possibility of the existence of trophectoderm-like hTSCs corresponding to earlier, preimplantation trophoblast cells.
Collapse
Affiliation(s)
- Moriyah Naama
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Yosef Buganim
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel.
| |
Collapse
|
68
|
Yang L, Liang P, Yang H, Coyne CB. Trophoblast organoids with physiological polarity model placental structure and function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.12.523752. [PMID: 36711688 PMCID: PMC9882188 DOI: 10.1101/2023.01.12.523752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Human trophoblast organoids (TOs) are a three-dimensional ex vivo culture model that can be used to study various aspects of placental development, physiology, and pathology. Previously, we showed that TOs derived from full-term human placental tissue could be used as models of trophoblast innate immune signaling and teratogenic virus infections. Here, we developed a method to culture TOs under conditions that recapitulate the cellular orientation of chorionic villi in vivo , with the multi-nucleated syncytiotrophoblast (STB) localized to the outer surface of organoids and the proliferative cytotrophoblasts (CTBs) located on the inner surface. We show that standard TOs containing the STB layer inside the organoid (STB in ) develop into organoids containing the STB on the outer surface (STB out ) when cultured in suspension with gentle agitation. STB out organoids secrete higher levels of select STB-associated hormones and cytokines, including human chorionic gonadotropin (hCG) and interferon (IFN)-λ2. Using membrane capacitance measurements, we also show that the outermost surface of STB out organoids contain large syncytia comprised of >50 nuclei compared to STB in organoids that contain small syncytia (<10 nuclei) and mononuclear cells. The growth of TOs under conditions that mimic the cellular orientation of chorionic villi in vivo thus allows for the study of a variety of aspects of placental biology under physiological conditions.
Collapse
Affiliation(s)
- Liheng Yang
- Department of Integrative Immunobiology, Duke University Medical Center, Durham, NC, USA
| | - Pengfei Liang
- Department of Biochemistry, Duke University Medical Center, Durham, NC, USA
| | - Huanghe Yang
- Department of Biochemistry, Duke University Medical Center, Durham, NC, USA
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA
| | - Carolyn B. Coyne
- Department of Integrative Immunobiology, Duke University Medical Center, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
69
|
Zhang Y, Wang H, Qiu P, Jiang J, Wu X, Mei J, Sun H. Decidual macrophages derived NO downregulates PD-L1 in trophoblasts leading to decreased Treg cells in recurrent miscarriage. Front Immunol 2023; 14:1180154. [PMID: 37520550 PMCID: PMC10379637 DOI: 10.3389/fimmu.2023.1180154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 06/26/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction Placental trophoblasts contribute to regulatory T (Treg) function via the programmed cell death-1 (PD-1)/PD-1 ligand 1 (PD-L1) pathway during normal pregnancy. Decreased expression of PD-L1 in trophoblasts was closely associated with Treg deficiency in the development of pregnancy failure. Thus, targeting PD-L1 might be a novel therapy to prevent pregnancy loss. However, the mechanisms for modulating the expression of PD-L1 in trophoblasts are an enigma. Methods The proportion of decidual Treg cells, and the profile of decidual macrophages (DMs) sampled from women with normal pregnancy (NP) and recurrent miscarriage (RM) were evaluated by flow cytometry. The expression of Yin and Yang 1 protein (YY1) and PD-L1 in human villous were measured by Immunohistochemistry (IHC), qRT-PCR and western blot. The determination of soluble PD-L1 (sPD-L1) in serum from NP and RM, and trophoblast conditioned media (TCM) was performed by the PD-L1 SimpleStep ELISA kit. Knockdown of YY1 was processed in the human trophoblast derived cell lines, HTR-8 and Bewo, with siYY1 transfection. Peripheral naïve CD4+ T cells were isolated from women with NP for the in vitro culture. The percentages of Treg cells differentiated from peripheral naïve CD4+ T cells were measured by flow cytometry. The interaction between YY1 and CD274 was proved by CHIP. The expression of inducible nitric oxide synthase (iNOS) in decidua was evaluated by IHC. The level of NO in serum from women with NP and RM was determined by the Griess reagent system. The effects of NO on YY1 were determined by the in vitro culture of HTR-8 cells with the NO donor, SNAP. The in vivo model comprising twelve pregnant mice and underwent different treatment. The percentages of Treg cells in murine uterus were measured by flow cytometry. Similarly, Western blot and IHC were performed to determine the expression of YY1 and PD-L1 in murine placenta. Results Decreased expression of YY1 and PD-L1 in trophoblasts and lower proportion of decidual Treg cells were observed in patients with RM. Knockdown of YY1 contributes to a lower expression of YY1 and PD-L1. Soluble PD-L1 in the supernatant from HTR-8 cells was also decreased with siYY1 administration. Lower Treg differentiation was observed in the presence of supernatant from HTR-8 cells treated with siYY1. CHIP analysis revealed that endogenous YY1 directly occupied the promoter region of the CD274 (PD-L1) gene. Accompanied with increased M1 DMs, higher NO was observed in serum sampled from patients with RM. In the presence of Reduced expression of YY1 and PD-L1 was observed in HTR-8 cells with the treatment of SNAP. Furthermore, less Treg differentiation was observed with SNAP treated TCM. Moreover, our in vivo data found that YY1 deficiency was associated with decreased PD-L1, which further resulting in less Treg differentiation and Treg deficiency at the maternal-fetal interface and increased embryo loss. Discussion Our work found the modulatory capacity of YY1 on PD-L1 in trophoblasts during early pregnancy. Furthermore, reduced YY1 was supposed resulting from higher levels of NO produced from the M1 DMs in RM.
Collapse
Affiliation(s)
| | | | | | | | | | - Jie Mei
- *Correspondence: Haixiang Sun, ; Jie Mei,
| | | |
Collapse
|
70
|
van Voorden AJ, Keijser R, Veenboer GJM, Lopes Cardozo SA, Diek D, Vlaardingerbroek JA, van Dijk M, Ris-Stalpers C, van Pelt AMM, Afink GB. EP300 facilitates human trophoblast stem cell differentiation. Proc Natl Acad Sci U S A 2023; 120:e2217405120. [PMID: 37406095 PMCID: PMC10334808 DOI: 10.1073/pnas.2217405120] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 06/05/2023] [Indexed: 07/07/2023] Open
Abstract
Early placenta development involves cytotrophoblast differentiation into extravillous trophoblast (EVT) and syncytiotrophoblast (STB). Defective trophoblast development and function may result in severe pregnancy complications, including fetal growth restriction and pre-eclampsia. The incidence of these complications is increased in pregnancies of fetuses affected by Rubinstein-Taybi syndrome, a developmental disorder predominantly caused by heterozygous mutations in CREB-binding protein (CREBBP) or E1A-binding protein p300 (EP300). Although the acetyltransferases CREBBP and EP300 are paralogs with many overlapping functions, the increased incidence of pregnancy complications is specific for EP300 mutations. We hypothesized that these complications have their origin in early placentation and that EP300 is involved in that process. Therefore, we investigated the role of EP300 and CREBBP in trophoblast differentiation, using human trophoblast stem cells (TSCs) and trophoblast organoids. We found that pharmacological CREBBP/EP300 inhibition blocks differentiation of TSCs into both EVT and STB lineages, and results in an expansion of TSC-like cells under differentiation-inducing conditions. Specific targeting by RNA interference or CRISPR/Cas9-mediated mutagenesis demonstrated that knockdown of EP300 but not CREBBP, inhibits trophoblast differentiation, consistent with the complications seen in Rubinstein-Taybi syndrome pregnancies. By transcriptome sequencing, we identified transforming growth factor alpha (TGFA, encoding TGF-α) as being strongly upregulated upon EP300 knockdown. Moreover, supplementing differentiation medium with TGF-α, which is a ligand for the epidermal growth factor receptor (EGFR), likewise affected trophoblast differentiation and resulted in increased TSC-like cell proliferation. These findings suggest that EP300 facilitates trophoblast differentiation by interfering with at least EGFR signaling, pointing towards a crucial role for EP300 in early human placentation.
Collapse
Affiliation(s)
- A. Jantine van Voorden
- Reproductive Biology Laboratory, Amsterdam Reproduction & Development, Amsterdam University Medical Centers, University of Amsterdam1105 AZ, Amsterdam, the Netherlands
| | - Remco Keijser
- Reproductive Biology Laboratory, Amsterdam Reproduction & Development, Amsterdam University Medical Centers, University of Amsterdam1105 AZ, Amsterdam, the Netherlands
| | - Geertruda J. M. Veenboer
- Reproductive Biology Laboratory, Amsterdam Reproduction & Development, Amsterdam University Medical Centers, University of Amsterdam1105 AZ, Amsterdam, the Netherlands
| | - Solange A. Lopes Cardozo
- Reproductive Biology Laboratory, Amsterdam Reproduction & Development, Amsterdam University Medical Centers, University of Amsterdam1105 AZ, Amsterdam, the Netherlands
| | - Dina Diek
- Reproductive Biology Laboratory, Amsterdam Reproduction & Development, Amsterdam University Medical Centers, University of Amsterdam1105 AZ, Amsterdam, the Netherlands
| | - Jennifer A. Vlaardingerbroek
- Reproductive Biology Laboratory, Amsterdam Reproduction & Development, Amsterdam University Medical Centers, University of Amsterdam1105 AZ, Amsterdam, the Netherlands
| | - Marie van Dijk
- Reproductive Biology Laboratory, Amsterdam Reproduction & Development, Amsterdam University Medical Centers, University of Amsterdam1105 AZ, Amsterdam, the Netherlands
| | - Carrie Ris-Stalpers
- Reproductive Biology Laboratory, Amsterdam Reproduction & Development, Amsterdam University Medical Centers, University of Amsterdam1105 AZ, Amsterdam, the Netherlands
- Department of Obstetrics and Gynaecology, Amsterdam Reproduction & Development, Amsterdam University Medical Centers, University of Amsterdam1105 AZ, Amsterdam, the Netherlands
| | - Ans M. M. van Pelt
- Reproductive Biology Laboratory, Amsterdam Reproduction & Development, Amsterdam University Medical Centers, University of Amsterdam1105 AZ, Amsterdam, the Netherlands
| | - Gijs B. Afink
- Reproductive Biology Laboratory, Amsterdam Reproduction & Development, Amsterdam University Medical Centers, University of Amsterdam1105 AZ, Amsterdam, the Netherlands
| |
Collapse
|
71
|
Milano-Foster J, Schulz LC. RISING STARS: Approaches to modeling placental function in preeclampsia in vitro and in vivo. J Endocrinol 2023; 258:e230008. [PMID: 37014303 PMCID: PMC10330201 DOI: 10.1530/joe-23-0008] [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: 03/21/2023] [Accepted: 04/04/2023] [Indexed: 04/05/2023]
Abstract
Modeling preeclampsia remains difficult due to the nature of the disease and the unique characteristics of the human placenta. Members of the Hominidae superfamily have a villous hemochorial placenta that is different in structure from those of other therian mammals, including the mouse hemochorial placenta, making this common animal model less ideal for studying this disease. Human placental tissues delivered from pregnancies complicated by preeclampsia are excellent for assessing the damage the disease causes but cannot answer how or when the disease begins. Symptoms of preeclampsia manifest halfway through pregnancy or later, making it currently impossible to identify preeclampsia in human tissues obtained from an early stage of pregnancy. Many animal and cell culture models recapitulate various aspects of preeclampsia, though none can on its own completely capture the complexity of human preeclampsia. It is particularly difficult to uncover the cause of the disease using models in which the disease is induced in the lab. However, the many ways by which preeclampsia-like features can be induced in a variety of laboratory animals are consistent with the idea that preeclampsia is a two-stage disease, in which a variety of initial insults may lead to placental ischemia, and ultimately systemic symptoms. The recent development of stem cell-based models, organoids, and various coculture systems have brought in vitro systems with human cells ever closer to recapitulating in vivo events that lead to placental ischemia.
Collapse
Affiliation(s)
- Jessica Milano-Foster
- Division of Animal Sciences, 245 Bond Life Sciences Center, 1201 Rollins Dr University of Missouri, Columbia MO 65211
| | - Laura C. Schulz
- Department of Obstetrics, Gynecology and Women’s Health, N610 Medical Sciences Building, Columbia, MO 65212
| |
Collapse
|
72
|
Elzinga FA, Khalili B, Touw DJ, Prins JR, Olinga P, Leuvenink HGD, van Goor H, Gordijn SJ, Nagelkerke A, Mian P. Placenta-on-a-Chip as an In Vitro Approach to Evaluate the Physiological and Structural Characteristics of the Human Placental Barrier upon Drug Exposure: A Systematic Review. J Clin Med 2023; 12:4315. [PMID: 37445348 DOI: 10.3390/jcm12134315] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/23/2023] [Accepted: 06/24/2023] [Indexed: 07/15/2023] Open
Abstract
Quantification of fetal drug exposure remains challenging since sampling from the placenta or fetus during pregnancy is too invasive. Currently existing in vivo (e.g., cord blood sampling) and ex vivo (e.g., placenta perfusion) models have inherent limitations. A placenta-on-a-chip model is a promising alternative. A systematic search was performed in PubMed on 2 February 2023, and Embase on 14 March 2023. Studies were included where placenta-on-a-chip was used to investigate placental physiology, placenta in different obstetric conditions, and/or fetal exposure to maternally administered drugs. Seventeen articles were included that used comparable approaches but different microfluidic devices and/or different cultured maternal and fetal cell lines. Of these studies, four quantified glucose transfer, four studies evaluated drug transport, three studies investigated nanoparticles, one study analyzed bacterial infection and five studies investigated preeclampsia. It was demonstrated that placenta-on-a-chip has the capacity to recapitulate the key characteristics of the human placental barrier. We aimed to identify knowledge gaps and provide the first steps towards an overview of current protocols for developing a placenta-on-a-chip, that facilitates comparison of results from different studies. Although models differ, they offer a promising approach for in vitro human placental and fetal drug studies under healthy and pathological conditions.
Collapse
Affiliation(s)
- Femke A Elzinga
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Behrad Khalili
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Daan J Touw
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
- Department of Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deunsinglaan 1, 9713 AV Groningen, The Netherlands
| | - Jelmer R Prins
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Peter Olinga
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deunsinglaan 1, 9713 AV Groningen, The Netherlands
| | - Henri G D Leuvenink
- Department of Surgery, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, Pathology Section, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Sanne J Gordijn
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Anika Nagelkerke
- Department of Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deunsinglaan 1, 9713 AV Groningen, The Netherlands
| | - Paola Mian
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
73
|
Naama M, Rahamim M, Zayat V, Sebban S, Radwan A, Orzech D, Lasry R, Ifrah A, Jaber M, Sabag O, Yassen H, Khatib A, Epsztejn-Litman S, Novoselsky-Persky M, Makedonski K, Deri N, Goldman-Wohl D, Cedar H, Yagel S, Eiges R, Buganim Y. Pluripotency-independent induction of human trophoblast stem cells from fibroblasts. Nat Commun 2023; 14:3359. [PMID: 37291192 PMCID: PMC10250329 DOI: 10.1038/s41467-023-39104-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 05/30/2023] [Indexed: 06/10/2023] Open
Abstract
Human trophoblast stem cells (hTSCs) can be derived from embryonic stem cells (hESCs) or be induced from somatic cells by OCT4, SOX2, KLF4 and MYC (OSKM). Here we explore whether the hTSC state can be induced independently of pluripotency, and what are the mechanisms underlying its acquisition. We identify GATA3, OCT4, KLF4 and MYC (GOKM) as a combination of factors that can generate functional hiTSCs from fibroblasts. Transcriptomic analysis of stable GOKM- and OSKM-hiTSCs reveals 94 hTSC-specific genes that are aberrant specifically in OSKM-derived hiTSCs. Through time-course-RNA-seq analysis, H3K4me2 deposition and chromatin accessibility, we demonstrate that GOKM exert greater chromatin opening activity than OSKM. While GOKM primarily target hTSC-specific loci, OSKM mainly induce the hTSC state via targeting hESC and hTSC shared loci. Finally, we show that GOKM efficiently generate hiTSCs from fibroblasts that harbor knockout for pluripotency genes, further emphasizing that pluripotency is dispensable for hTSC state acquisition.
Collapse
Affiliation(s)
- Moriyah Naama
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, 91120, Jerusalem, Israel
| | - Moran Rahamim
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, 91120, Jerusalem, Israel
| | - Valery Zayat
- Department of Stem Cell Bioengineering, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, 02-106, Poland
| | - Shulamit Sebban
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, 91120, Jerusalem, Israel
| | - Ahmed Radwan
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, 91120, Jerusalem, Israel
| | - Dana Orzech
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, 91120, Jerusalem, Israel
| | - Rachel Lasry
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, 91120, Jerusalem, Israel
| | - Annael Ifrah
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, 91120, Jerusalem, Israel
| | - Mohammad Jaber
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, 91120, Jerusalem, Israel
| | - Ofra Sabag
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, 91120, Jerusalem, Israel
| | - Hazar Yassen
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, 91120, Jerusalem, Israel
| | - Areej Khatib
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, 91120, Jerusalem, Israel
| | - Silvina Epsztejn-Litman
- Stem Cell Research Laboratory, Medical Genetics Institute, Shaare Zedek Medical Center, 91031, Jerusalem, Israel
- The Hebrew University School of Medicine, 91120, Jerusalem, Israel
| | - Michal Novoselsky-Persky
- The Magda and Richard Hoffman Laboratory of Human Placental Research, Department of Obstetrics and Gynecology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Kirill Makedonski
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, 91120, Jerusalem, Israel
| | - Noy Deri
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, 91120, Jerusalem, Israel
| | - Debra Goldman-Wohl
- The Magda and Richard Hoffman Laboratory of Human Placental Research, Department of Obstetrics and Gynecology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Howard Cedar
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, 91120, Jerusalem, Israel
| | - Simcha Yagel
- The Magda and Richard Hoffman Laboratory of Human Placental Research, Department of Obstetrics and Gynecology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Rachel Eiges
- Stem Cell Research Laboratory, Medical Genetics Institute, Shaare Zedek Medical Center, 91031, Jerusalem, Israel
- The Hebrew University School of Medicine, 91120, Jerusalem, Israel
| | - Yosef Buganim
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, 91120, Jerusalem, Israel.
| |
Collapse
|
74
|
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
|
75
|
Karakis V, Jabeen M, Britt JW, Cordiner A, Mischler A, Li F, San Miguel A, Rao BM. Laminin switches terminal differentiation fate of human trophoblast stem cells under chemically defined culture conditions. J Biol Chem 2023; 299:104650. [PMID: 36972789 PMCID: PMC10176266 DOI: 10.1016/j.jbc.2023.104650] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 03/16/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Human trophoblast stem cells (hTSCs) have emerged as a powerful tool to model early placental development in vitro. Analogous to the epithelial cytotrophoblast in the placenta, hTSCs can differentiate into cells of the extravillous trophoblast (EVT) lineage or the multinucleate syncytiotrophoblast (STB). Here we present a chemically defined culture system for STB and EVT differentiation of hTSCs. Notably, in contrast to current approaches, we neither utilize forskolin for STB formation nor transforming growth factor-beta (TGFβ) inhibitors or a passage step for EVT differentiation. Strikingly, the presence of a single additional extracellular cue-laminin-111-switched the terminal differentiation of hTSCs from STB to the EVT lineage under these conditions. In the absence of laminin-111, STB formation occurred, with cell fusion comparable to that obtained with differentiation mediated by forskolin; however, in the presence of laminin-111, hTSCs differentiated to the EVT lineage. Protein expression of nuclear hypoxia-inducible factors (HIF1α and HIF2α) was upregulated during EVT differentiation mediated by laminin-111 exposure. A heterogeneous mixture of Notch1+ EVTs in colonies and HLA-G+ single-cell EVTs were obtained without a passage step, reminiscent of heterogeneity in vivo. Further analysis showed that inhibition of TGFβ signaling affected both STB and EVT differentiation mediated by laminin-111 exposure. TGFβ inhibition during EVT differentiation resulted in decreased HLA-G expression and increased Notch1 expression. On the other hand, TGFβ inhibition prevented STB formation. The chemically defined culture system for hTSC differentiation established herein facilitates quantitative analysis of heterogeneity that arises during hTSC differentiation and will enable mechanistic studies in vitro.
Collapse
Affiliation(s)
- Victoria Karakis
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Mahe Jabeen
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - John W Britt
- Department of Genetics, North Carolina State University, Raleigh, North Carolina, USA
| | - Abigail Cordiner
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Adam Mischler
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Feng Li
- Department of Pathology and Laboratory Medicine, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
| | - Adriana San Miguel
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Balaji M Rao
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA; Golden LEAF Biomanufacturing Training and Education Center, North Carolina State University, Raleigh, North Carolina, USA.
| |
Collapse
|
76
|
Haider S, Beristain AG. Human organoid systems in modeling reproductive tissue development, function, and disease. Hum Reprod 2023:7147082. [PMID: 37119533 DOI: 10.1093/humrep/dead085] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 04/12/2023] [Indexed: 05/01/2023] Open
Abstract
Research focused on human reproductive biology has primarily relied upon clinical samples affording mainly descriptive studies with limited implementation of functional or mechanistic understanding. More importantly, restricted access to human embryonic material has necessitated the use of animals, primarily rats and mice, and short-term primary cell cultures derived from human patient material. While reproductive developmental processes are generally conserved across mammals, specific features unique to human reproduction have resulted in the development of human-based in vitro systems designed to retain or recapitulate key molecular and cellular processes important in humans. Of note, major advances in 3D epithelial stem cell-based systems modeling human reproductive organ development have been made. These cultures, broadly referred to as organoids, enable research aimed at understanding cellular hierarchies and processes controlling cellular differentiation and function. Moreover, organoids allow the pre-clinical testing of pharmacological substances, both from safety and efficacy standpoints, and hold large potential in driving aspects of personalized medicine that were previously not possible with traditional models. In this mini-review, we focus on summarizing the current state of regenerative organoid culture systems of the female and male reproductive tracts that model organ development, maintenance, and function. Specifically, we will introduce stem cell-based organoid models of the ovary/fallopian tube, endometrium, cervix, prostate gland, and testes. We will also describe organoid systems of the pre-implanting blastocyst and trophoblast, as the blastocyst and its extraembryonic trophectoderm are central to fetal, maternal, and overall pregnancy health. We describe the foundational studies leading to their development and outline the utility as well as specific limitations that are unique and common to many of these in vitro platforms.
Collapse
Affiliation(s)
- Sandra Haider
- Department of Obstetrics & Gynecology, Medical University of Vienna, Vienna, Austria
| | - 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
|
77
|
Burton GJ, Jauniaux E. The human placenta: new perspectives on its formation and function during early pregnancy. Proc Biol Sci 2023; 290:20230191. [PMID: 37072047 PMCID: PMC10113033 DOI: 10.1098/rspb.2023.0191] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2023] Open
Abstract
The placenta has evolved to support the development of the embryo and fetus during the different intrauterine periods of life. By necessity, its development must precede that of the embryo. There is now evidence that during embryogenesis and organogenesis, the development of the human placenta is supported by histotrophic nutrition secreted from endometrial glands rather than maternal blood. These secretions provide a plentiful supply of glucose, lipids, glycoproteins and growth factors that stimulate rapid proliferation and differentiation of the villous trophoblast. Furthermore, evidence from endometrial gland organoids indicates that expression and secretion of these products are upregulated following sequential exposure to oestrogen, progesterone and trophoblastic and decidual hormones, in particular prolactin. Hence, a feed-forward signalling dialogue is proposed among the trophoblast, decidua and glands that enables the placenta to stimulate its own development, independent of that of the embryo. Many common complications of pregnancy represent a spectrum of disorders associated with deficient trophoblast proliferation. Increasing evidence suggests that this spectrum is mirrored by one of impaired decidualization, potentially compromising histotroph secretion through diminished prolactin secretion and reduced gland function. Optimizing endometrial wellbeing prior to conception may therefore help to prevent common pregnancy complications, such as miscarriage, growth restriction and pre-eclampsia.
Collapse
Affiliation(s)
- Graham J Burton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Eric Jauniaux
- EGA Institute for Women's Health, Faculty of Population Health Sciences, University College London, London, UK
| |
Collapse
|
78
|
Arutyunyan A, Roberts K, Troulé K, Wong FCK, Sheridan MA, Kats I, Garcia-Alonso L, Velten B, Hoo R, Ruiz-Morales ER, Sancho-Serra C, Shilts J, Handfield LF, Marconato L, Tuck E, Gardner L, Mazzeo CI, Li Q, Kelava I, Wright GJ, Prigmore E, Teichmann SA, Bayraktar OA, Moffett A, Stegle O, Turco MY, Vento-Tormo R. Spatial multiomics map of trophoblast development in early pregnancy. Nature 2023; 616:143-151. [PMID: 36991123 PMCID: PMC10076224 DOI: 10.1038/s41586-023-05869-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 02/21/2023] [Indexed: 03/31/2023]
Abstract
The relationship between the human placenta-the extraembryonic organ made by the fetus, and the decidua-the mucosal layer of the uterus, is essential to nurture and protect the fetus during pregnancy. Extravillous trophoblast cells (EVTs) derived from placental villi infiltrate the decidua, transforming the maternal arteries into high-conductance vessels1. Defects in trophoblast invasion and arterial transformation established during early pregnancy underlie common pregnancy disorders such as pre-eclampsia2. Here we have generated a spatially resolved multiomics single-cell atlas of the entire human maternal-fetal interface including the myometrium, which enables us to resolve the full trajectory of trophoblast differentiation. We have used this cellular map to infer the possible transcription factors mediating EVT invasion and show that they are preserved in in vitro models of EVT differentiation from primary trophoblast organoids3,4 and trophoblast stem cells5. We define the transcriptomes of the final cell states of trophoblast invasion: placental bed giant cells (fused multinucleated EVTs) and endovascular EVTs (which form plugs inside the maternal arteries). We predict the cell-cell communication events contributing to trophoblast invasion and placental bed giant cell formation, and model the dual role of interstitial EVTs and endovascular EVTs in mediating arterial transformation during early pregnancy. Together, our data provide a comprehensive analysis of postimplantation trophoblast differentiation that can be used to inform the design of experimental models of the human placenta in early pregnancy.
Collapse
Affiliation(s)
- Anna Arutyunyan
- Wellcome Sanger Institute, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | | | | | | | - Megan A Sheridan
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Ilia Kats
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Britta Velten
- Wellcome Sanger Institute, Cambridge, UK
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Regina Hoo
- Wellcome Sanger Institute, Cambridge, UK
| | | | | | | | | | - Luca Marconato
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | | | - Lucy Gardner
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | | | - Qian Li
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Iva Kelava
- Wellcome Sanger Institute, Cambridge, UK
| | - Gavin J Wright
- Department of Biology, Hull York Medical School, York Biomedical Research Institute, University of York, York, UK
| | | | - Sarah A Teichmann
- Wellcome Sanger Institute, Cambridge, UK
- Theory of Condensed Matter, Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, UK
| | | | - Ashley Moffett
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
- Department of Pathology, University of Cambridge, Cambridge, UK.
| | - Oliver Stegle
- Wellcome Sanger Institute, Cambridge, UK.
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany.
| | - Margherita Y Turco
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
- Department of Pathology, University of Cambridge, Cambridge, UK.
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland.
| | - Roser Vento-Tormo
- Wellcome Sanger Institute, Cambridge, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| |
Collapse
|
79
|
Fantone S, Ermini L, Piani F, Di Simone N, Barbaro G, Giannubilo SR, Gesuita R, Tossetta G, Marzioni D. Downregulation of argininosuccinate synthase 1 (ASS1) is associated with hypoxia in placental development. Hum Cell 2023; 36:1190-1198. [PMID: 36995581 DOI: 10.1007/s13577-023-00901-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/24/2023] [Indexed: 03/31/2023]
Abstract
Argininosuccinate synthase (ASS1) is involved in nitric oxide production, which has a key role in placental development improving pregnancy outcomes. Syncytiotrophoblast and extravillous trophoblast differentiations are milestones of placental development and their impairment can cause pathologies, such as preeclampsia (PE) and fetal growth restriction (FGR). Immunohistochemistry and Western blotting were used to localize and quantify ASS1 in first trimester (8.2 ± 1.8 weeks), third trimester (38.6 ± 1.1 weeks), and PE (36.3 ± 1.5 weeks) placentas. In addition, cell cultures were used to evaluate ASS1 expression under hypoxic conditions and the syncytialization process. Our data showed that ASS1 is localized in the villous cytotrophoblast of first trimester, third trimester, and PE placentas, while the villous cytotrophoblast adjacent to the extravillous trophoblast of cell columns as well as the extravillous trophoblast were negative for ASS1 in first trimester placentas. In addition, ASS1 was decreased in third trimester compared to the first trimester placentas (p = 0.003) and no differences were detected between third trimester and PE placentas. Moreover, ASS1 expression was decreased in hypoxic conditions and syncytialized cells compared to those not syncytialized. In conclusion, we suggest that the expression of ASS1 in villous cytotrophoblast is related to maintaining proliferative phenotype, while ASS1 absence may be involved in promoting the differentiation of villous cytotrophoblast in extravillous cytotrophoblast of cell columns in first trimester placentas.
Collapse
Affiliation(s)
- Sonia Fantone
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126, Ancona, Italy
| | - Leonardo Ermini
- Department of Life Science, University of Siena, 53100, Siena, Italy
| | - Federica Piani
- Cardiovascular Internal Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40128, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, 40138, Bologna, Italy
| | - Nicoletta Di Simone
- Department of Biomedical Sciences, Humanitas University, 20072, Milan, Italy
- IRCCS Humanitas Research Hospital, 20089, Milan, Italy
| | - Greta Barbaro
- Istituto di Clinica Ostetrica e Ginecologica, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Stefano Raffaele Giannubilo
- Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, Azienda Ospedaliero Universitaria, 60126, Ancona, Italy
| | - Rosaria Gesuita
- Centre of Epidemiology and Biostatistics, Università Politecnica delle Marche, 60126, Ancona, Italy
| | - Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126, Ancona, Italy.
- Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, Azienda Ospedaliero Universitaria, 60126, Ancona, Italy.
| | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126, Ancona, Italy
| |
Collapse
|
80
|
Zhuang BM, Cao DD, Liu XF, Wang L, Lin XL, Duan YG, Lee CL, Chiu PCN, Yeung WSB, Yao YQ. Application of a JEG-3 organoid model to study HLA-G function in the trophoblast. Front Immunol 2023; 14:1130308. [PMID: 37006248 PMCID: PMC10050466 DOI: 10.3389/fimmu.2023.1130308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 02/28/2023] [Indexed: 03/17/2023] Open
Abstract
The human placenta is a unique temporary organ with a mysterious immune tolerance. The formation of trophoblast organoids has advanced the study of placental development. HLA-G is uniquely expressed in the extravillous trophoblast (EVT) and has been linked to placental disorders. With older experimental methodologies, the role of HLA-G in trophoblast function beyond immunomodulation is still contested, as is its role during trophoblast differentiation. Organoid models incorporating CRISPR/Cas9 technology were used to examine the role of HLA-G in trophoblast function and differentiation. JEG-3 trophoblast organoids (JEG-3-ORGs) were established that highly expressed trophoblast representative markers and had the capacity to differentiate into EVT. CRISPR/Cas9 based on HLA-G knockout (KO) significantly altered the trophoblast immunomodulatory effect on the cytotoxicity of natural killer cells, as well as the trophoblast regulatory effect on HUVEC angiogenesis, but had no effect on the proliferation and invasion of JEG-3 cells and the formation of TB-ORGs. RNA-sequencing analysis further demonstrated that JEG-3 KO cells followed similar biological pathways as their wild-type counterparts during the formation of TB-ORGs. In addition, neither HLA-G KO nor the exogenous addition of HLA-G protein during EVT differentiation from JEG-3-ORGs altered the temporal expression of the known EVT marker genes. Based on the JEG-3 KO (disruption of exons 2 and 3) cell line and the TB-ORGs model, it was determined that HLA-G has a negligible effect on trophoblast invasion and differentiation. Despite this, JEG-3-ORG remains a valuable model for studying trophoblast differentiation.
Collapse
Affiliation(s)
- Bai-Mei Zhuang
- Medical School of Chinese People’s Liberation Army, Chinese People’s Liberation Army General Hospital, Beijing, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Shenzhen, Guangdong, China
| | - Dan-Dan Cao
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Shenzhen, Guangdong, China
| | - Xiao-Feng Liu
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Shenzhen, Guangdong, China
| | - Li Wang
- Department of Obstetrics and Gynecology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Xiao-Li Lin
- Department of Obstetrics and Gynecology, Affiliated Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Yong-Gang Duan
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Shenzhen, Guangdong, China
| | - Cheuk-Lun Lee
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Shenzhen, Guangdong, China
- Department of Obstetrics and Gynecology, HKU Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Philip C. N. Chiu
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Shenzhen, Guangdong, China
- Department of Obstetrics and Gynecology, HKU Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - William S. B. Yeung
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Shenzhen, Guangdong, China
- Department of Obstetrics and Gynecology, HKU Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- *Correspondence: William S. B. Yeung, ; Yuan-Qing Yao,
| | - Yuan-Qing Yao
- Medical School of Chinese People’s Liberation Army, Chinese People’s Liberation Army General Hospital, Beijing, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Shenzhen, Guangdong, China
- Department of Obstetrics and Gynecology, The First Medical Centre, Chinese People’s Liberation Army General Hospital, Beijing, China
- *Correspondence: William S. B. Yeung, ; Yuan-Qing Yao,
| |
Collapse
|
81
|
Mohammad S, Bhattacharjee J, Tzaneva V, Hutchinson KA, Shaikh M, Fernandes da Silva D, Burger D, Adamo KB. The Influence of Exercise-Associated Small Extracellular Vesicles on Trophoblasts In Vitro. Biomedicines 2023; 11:biomedicines11030857. [PMID: 36979835 PMCID: PMC10045992 DOI: 10.3390/biomedicines11030857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/01/2023] [Accepted: 03/09/2023] [Indexed: 03/15/2023] Open
Abstract
Exercise induces the release of small extracellular vesicles (sEVs) into circulation that are postulated to mediate tissue cross-talk during exercise. We previously reported that pregnant individuals released greater levels of sEVs into circulation after exercise compared to matched non-pregnant controls, but their biological functions remain unknown. In this study, sEVs isolated from the plasma of healthy pregnant and non-pregnant participants after a single bout of moderate-intensity exercise were evaluated for their impact on trophoblasts in vitro. Exercise-associated sEVs were found localized within the cytoplasm of BeWo choriocarcinoma cells, used to model trophoblasts in vitro. Exposure to exercise-associated sEVs did not significantly alter BeWo cell proliferation, gene expression of angiogenic growth factors VEGF and PLGF, or the release of the hormone human chorionic gonadotropin. The results from this pilot study support that exercise-associated sEVs could interact with trophoblasts in vitro, and warrant further investigation to reveal their potential role in communicating the effects of exercise to the maternal–fetal interface.
Collapse
Affiliation(s)
- Shuhiba Mohammad
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Jayonta Bhattacharjee
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Velislava Tzaneva
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Kelly Ann Hutchinson
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Madeeha Shaikh
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Danilo Fernandes da Silva
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Dylan Burger
- Kidney Research Centre, Department of Cellular and Molecular Medicine, The Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON K1H 8L6, Canada
| | - Kristi B. Adamo
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
- Correspondence:
| |
Collapse
|
82
|
Faral-Tello P, Pagotto R, Bollati-Fogolín M, Francia ME. Modeling the human placental barrier to understand Toxoplasma gondii´s vertical transmission. Front Cell Infect Microbiol 2023; 13:1130901. [PMID: 36968102 PMCID: PMC10034043 DOI: 10.3389/fcimb.2023.1130901] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/23/2023] [Indexed: 03/11/2023] Open
Abstract
Toxoplasma gondii is a ubiquitous apicomplexan parasite that can infect virtually any warm-blooded animal. Acquired infection during pregnancy and the placental breach, is at the core of the most devastating consequences of toxoplasmosis. T. gondii can severely impact the pregnancy’s outcome causing miscarriages, stillbirths, premature births, babies with hydrocephalus, microcephaly or intellectual disability, and other later onset neurological, ophthalmological or auditory diseases. To tackle T. gondii’s vertical transmission, it is important to understand the mechanisms underlying host-parasite interactions at the maternal-fetal interface. Nonetheless, the complexity of the human placenta and the ethical concerns associated with its study, have narrowed the modeling of parasite vertical transmission to animal models, encompassing several unavoidable experimental limitations. Some of these difficulties have been overcome by the development of different human cell lines and a variety of primary cultures obtained from human placentas. These cellular models, though extremely valuable, have limited ability to recreate what happens in vivo. During the last decades, the development of new biomaterials and the increase in stem cell knowledge have led to the generation of more physiologically relevant in vitro models. These cell cultures incorporate new dimensions and cellular diversity, emerging as promising tools for unraveling the poorly understood T. gondii´s infection mechanisms during pregnancy. Herein, we review the state of the art of 2D and 3D cultures to approach the biology of T. gondii pertaining to vertical transmission, highlighting the challenges and experimental opportunities of these up-and-coming experimental platforms.
Collapse
Affiliation(s)
- Paula Faral-Tello
- Laboratory of Apicomplexan Biology, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Romina Pagotto
- Cell Biology Unit, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | | | - Maria E. Francia
- Laboratory of Apicomplexan Biology, Institut Pasteur de Montevideo, Montevideo, Uruguay
- Departamento de Parasitología y Micología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- *Correspondence: Maria E. Francia,
| |
Collapse
|
83
|
Dietrich B, Kunihs V, Pollheimer J, Knöfler M, Haider S. 3D organoid formation and EVT differentiation of various trophoblastic cell lines. Placenta 2023; 133:19-22. [PMID: 36696785 DOI: 10.1016/j.placenta.2023.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/13/2022] [Accepted: 01/07/2023] [Indexed: 01/15/2023]
Abstract
3-dimensional trophoblast organoids (TB-ORG) represent a reliable model for studying extravillous trophoblast (EVT) lineage formation and differentiation. However, restricted access to first trimester placentae requires alternative cell sources for establishing placental organoids. Recently, we demonstrated EVT differentiation in JEG-3-derived organoids. Consequently, we herein tested whether other commonly used trophoblastic cell lines, ACH-3P, HTR-8/SVneo, and SWAN-71 were capable of self-organizing into organoids and subsequent EVT differentiation. Notably, only ACH-3P formed organoids under stemness conditions mimicking TB-ORG architectures, and induction of EVT differentiation provoked formation of HLA-Gpos areas. Hence ACH-3P-ORGs provide another organoid model for studying controlled EVT lineage formation and differentiation.
Collapse
Affiliation(s)
- Bianca Dietrich
- Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Placental Development Group, Medical University of Vienna, Vienna, Austria
| | - Victoria Kunihs
- Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Placental Development Group, Medical University of Vienna, Vienna, Austria
| | - Jürgen Pollheimer
- Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Maternal-Fetal Immunology Group, Medical University of Vienna, Vienna, Austria
| | - Martin Knöfler
- Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Placental Development Group, Medical University of Vienna, Vienna, Austria
| | - Sandra Haider
- Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Placental Development Group, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
84
|
Calà G, Sina B, De Coppi P, Giobbe GG, Gerli MFM. Primary human organoids models: Current progress and key milestones. Front Bioeng Biotechnol 2023; 11:1058970. [PMID: 36959902 PMCID: PMC10029057 DOI: 10.3389/fbioe.2023.1058970] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 02/22/2023] [Indexed: 03/06/2023] Open
Abstract
During the past 10 years the world has experienced enormous progress in the organoids field. Human organoids have shown huge potential to study organ development, homeostasis and to model diseases in vitro. The organoid technology has been widely and increasingly applied to generate patient-specific in vitro 3D cultures, starting from both primary and reprogrammed stem/progenitor cells. This has consequently fostered the development of innovative disease models and new regenerative therapies. Human primary, or adult stem/progenitor cell-derived, organoids can be derived from both healthy and pathological primary tissue samples spanning from fetal to adult age. The resulting 3D culture can be maintained for several months and even years, while retaining and resembling its original tissue's properties. As the potential of this technology expands, new approaches are emerging to further improve organoid applications in biology and medicine. This review discusses the main organs and tissues which, as of today, have been modelled in vitro using primary organoid culture systems. Moreover, we also discuss the advantages, limitations, and future perspectives of primary human organoids in the fields of developmental biology, disease modelling, drug testing and regenerative medicine.
Collapse
Affiliation(s)
- Giuseppe Calà
- Division of Surgery and Interventional Science, Department of Surgical Biotechnology, University College London, London, United Kingdom
- Stem Cell and Regenerative Medicine Section, Zayed Centre for Research into Rare Disease in Children, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Beatrice Sina
- Division of Surgery and Interventional Science, Department of Surgical Biotechnology, University College London, London, United Kingdom
- Politecnico di Milano, Milano, Italy
| | - Paolo De Coppi
- Stem Cell and Regenerative Medicine Section, Zayed Centre for Research into Rare Disease in Children, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
- Specialist Neonatal and Paediatric Surgery, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Giovanni Giuseppe Giobbe
- Stem Cell and Regenerative Medicine Section, Zayed Centre for Research into Rare Disease in Children, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Mattia Francesco Maria Gerli
- Division of Surgery and Interventional Science, Department of Surgical Biotechnology, University College London, London, United Kingdom
- Stem Cell and Regenerative Medicine Section, Zayed Centre for Research into Rare Disease in Children, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
85
|
Motomura K, Hara M, Ito I, Morita H, Matsumoto K. Roles of human trophoblasts' pattern recognition receptors in host defense and pregnancy complications. J Reprod Immunol 2023; 156:103811. [PMID: 36669386 DOI: 10.1016/j.jri.2023.103811] [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: 08/03/2022] [Revised: 12/24/2022] [Accepted: 01/12/2023] [Indexed: 01/14/2023]
Abstract
The immune system in pregnancy is able to protect pregnant mothers and fetuses from pathogenic microorganisms even while permitting the mother to tolerate the semi-allogenic fetus. Trophoblasts, which are fetal-derived placental cells, play a central role on both sides of this duality at the maternal-fetal interface. In brief, the trophoblasts express pattern recognition receptors (PRRs) and are involved in the local innate immune response in the placenta. That response eliminates pathogenic microbes but also causes tissue damage. In this review, we summarize the research findings to date regarding the roles of those human trophoblast PRRs. Multiple types of PRRs (Toll-like receptors, Nod-like receptors, and RIG-I-like receptors) are expressed in the placenta and on trophoblasts. Trophoblasts' PRRs participate in protecting the fetus against viruses, bacteria, and parasites by triggering production of proinflammatory cytokines and chemokines in the placenta. On the negative side, PRR signaling in trophoblasts can also initiate inflammation and trophoblast cell death, which can lead to placental inflammation-associated pregnancy complications such as preeclampsia, anti-phospholipid antibody syndrome, and miscarriage. Further elucidation of these dual roles of trophoblasts' PRRs may shed light on the mechanisms by which fetuses are protected against congenital infections and also give us a better understanding of the etiologies of pregnancy complications, which can help us prevent/reduce adverse prenatal/neonatal outcomes.
Collapse
Affiliation(s)
- Kenichiro Motomura
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan; Center for Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Tokyo 157-8535, Japan.
| | - Mariko Hara
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan; Department of Otorhinolaryngology, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Ikuyo Ito
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan; Department of Pediatrics, School of Medicine, Yokohama City University, Kanagawa 236-0004, Japan
| | - Hideaki Morita
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan; Allergy Center, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Kenji Matsumoto
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan.
| |
Collapse
|
86
|
Koltsova EM, Martyanov AA, Podoplelova NA. Procoagulant Properties of Extracellular Vesicles in Normal and Pathological Pregnancy. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES A: MEMBRANE AND CELL BIOLOGY 2023. [DOI: 10.1134/s1990747822060071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
|
87
|
Burgaud M, Bretin B, Reignier A, De Vos J, David L. [New models to study human embryonic development]. Med Sci (Paris) 2023; 39:129-136. [PMID: 36799747 DOI: 10.1051/medsci/2023018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023] Open
Abstract
Since 2021, assisted reproductive technologies (ART) are available to infertile couples, but also to single women and female couples. The process of in vitro fertilization (IVF) has allowed to cross the threshold of 5 million births worldwide, between 1978 and 2013. However, the failure rate per each IVF cycle is estimated to be around 75%. Therefore, there is a need to better understand human embryonic development in order to improve the success rate of IVF. Study models have evolved significantly in recent years: development of embryo culture, sequencing of the transcriptome of individualized cells, discovery of culture conditions for naive pluripotent stem cells and generation of blastoids. Here, we review these recent advances in human embryo modeling that establish a new knowledge base for improving ART.
Collapse
Affiliation(s)
- Mathilde Burgaud
- Nantes université, CHU Nantes, Inserm, CR2TI, F-44000 Nantes, France
| | - Betty Bretin
- Nantes université, CHU Nantes, Inserm, CR2TI, F-44000 Nantes, France
| | - Arnaud Reignier
- Nantes université, CHU Nantes, Inserm, CR2TI, F-44000 Nantes, France - CHU Nantes, Service de biologie de la reproduction, F-44000 Nantes, France
| | - John De Vos
- IRMB, Univ Montpellier, Inserm, CHU Montpellier, Montpellier, France
| | - Laurent David
- Nantes université, CHU Nantes, Inserm, CR2TI, F-44000 Nantes, France - Nantes université, CHU Nantes, Inserm, CNRS, BioCore, F-44000 Nantes, France
| |
Collapse
|
88
|
Krausová M, Braun D, Buerki-Thurnherr T, Gundacker C, Schernhammer E, Wisgrill L, Warth B. Understanding the Chemical Exposome During Fetal Development and Early Childhood: A Review. Annu Rev Pharmacol Toxicol 2023; 63:517-540. [PMID: 36202091 DOI: 10.1146/annurev-pharmtox-051922-113350] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Early human life is considered a critical window of susceptibility to external exposures. Infants are exposed to a multitude of environmental factors, collectively referred to as the exposome. The chemical exposome can be summarized as the sum of all xenobiotics that humans are exposed to throughout a lifetime. We review different exposure classes and routes that impact fetal and infant metabolism and the potential toxicological role of mixture effects. We also discuss the progress in human biomonitoring and present possiblemodels for studying maternal-fetal transfer. Data gaps on prenatal and infant exposure to xenobiotic mixtures are identified and include natural biotoxins, in addition to commonly reported synthetic toxicants, to obtain a more holistic assessment of the chemical exposome. We highlight the lack of large-scale studies covering a broad range of xenobiotics. Several recommendations to advance our understanding of the early-life chemical exposome and the subsequent impact on health outcomes are proposed.
Collapse
Affiliation(s)
- Magdaléna Krausová
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria; , ,
| | - Dominik Braun
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria; , ,
| | - Tina Buerki-Thurnherr
- Swiss Federal Laboratories for Materials Science and Technology, Laboratory for Particles Biology Interactions, St. Gallen, Switzerland;
| | - Claudia Gundacker
- Center for Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria; .,Exposome Austria, Research Infrastructure and National EIRENE Hub, Austria
| | - Eva Schernhammer
- Exposome Austria, Research Infrastructure and National EIRENE Hub, Austria.,Center for Public Health, Department of Epidemiology, Medical University of Vienna, Vienna, Austria; .,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
| | - Lukas Wisgrill
- Exposome Austria, Research Infrastructure and National EIRENE Hub, Austria.,Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria;
| | - Benedikt Warth
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria; , , .,Exposome Austria, Research Infrastructure and National EIRENE Hub, Austria
| |
Collapse
|
89
|
Matsumoto S, Okamura E, Muto M, Ema M. Similarities and differences in placental development between humans and cynomolgus monkeys. Reprod Med Biol 2023; 22:e12522. [PMID: 37377753 PMCID: PMC10292683 DOI: 10.1002/rmb2.12522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/07/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Background The placenta is an extraembryonic organ, which is essential to maintain a normal pregnancy. However, placental development in humans is poorly understood because of technical and ethical reasons. Methods We analyzed the anatomical localization of each trophoblastic subtype in the cynomolgus monkey placenta by immunohistochemistry in the early second trimester. Histological differences among the mouse, cynomolgus monkey, and human placenta were compared. The PubMed database was used to search for studies on placentation in rodents and primates. Main findings The anatomical structures and subtypes of the placenta in cynomolgus monkeys are highly similar to those in humans, with the exception of fewer interstitial extravillous trophoblasts in cynomolgus monkeys. Conclusion The cynomolgus monkey appears to be a good animal model to investigate human placentation.
Collapse
Affiliation(s)
- Shoma Matsumoto
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life ScienceShiga University of Medical ScienceOtsuJapan
| | - Eiichi Okamura
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life ScienceShiga University of Medical ScienceOtsuJapan
| | - Masanaga Muto
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life ScienceShiga University of Medical ScienceOtsuJapan
| | - Masatsugu Ema
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life ScienceShiga University of Medical ScienceOtsuJapan
- Institute for the Advanced Study of Human Biology (WPI‐ASHBi)Kyoto UniversityKyotoJapan
| |
Collapse
|
90
|
Cui K, Chen T, Zhu Y, Shi Y, Guo Y, Qin J. Engineering placenta-like organoids containing endogenous vascular cells from human-induced pluripotent stem cells. Bioeng Transl Med 2023; 8:e10390. [PMID: 36684087 PMCID: PMC9842056 DOI: 10.1002/btm2.10390] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 07/17/2022] [Accepted: 07/27/2022] [Indexed: 01/25/2023] Open
Abstract
The placenta is an essential organ that maintains the health of both the fetus and its mother. Understanding the development of human placenta has been hindered by the limitations of existing animal models and monolayer cell cultures. Models that can recapitulate the essential aspects of human placental multicellular components and vasculature are still lacking. Herein, we presented a new strategy to establish placenta-like organoids with vascular-like structures from human-induced pluripotent stem cells in a defined three-dimensional (3D) culture system. The resulting placenta-like tissue resembles first-trimester human placental development in terms of complex placental components and secretory function. The multicellular tissue was characterized by the inclusion of trophoblasts (cytotrophoblasts, syncytiotrophoblasts, extravillous trophoblasts, and other endogenous vascular cells), which were identified by immunofluorescence, flow cytometry analyses, real-time quantitative reverse transcription polymerase chain reaction and single-cell RNA-seq. Moreover, the 3D tissue was able to secrete the placenta-specific hormone human chorionic gonadotropin β (hCG-β) and vascular endothelial growth factor A (VEGFA). The tissue responded to the inflammatory factor tumor necrosis factor-α (TNF-α) and VEGF receptor inhibitors. This new model system can represent the major features of placental cellular components, and function, which have not been realized in 2D monolayer cultures. The developed tissue system might open new avenues for studying normal early human placental development and its disease states.
Collapse
Affiliation(s)
- Kangli Cui
- Division of Biotechnology, CAS Key Laboratory of SSACDalian Institute of Chemical Physics, Chinese Academy of SciencesDalianChina
- University of Chinese Academy of SciencesBeijingChina
| | - Tingwei Chen
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
| | - Yujuan Zhu
- Division of Biotechnology, CAS Key Laboratory of SSACDalian Institute of Chemical Physics, Chinese Academy of SciencesDalianChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yang Shi
- Dalian Municipal Woman and Children's Medical CenterDalianChina
| | - Yaqiong Guo
- Division of Biotechnology, CAS Key Laboratory of SSACDalian Institute of Chemical Physics, Chinese Academy of SciencesDalianChina
- University of Chinese Academy of SciencesBeijingChina
| | - Jianhua Qin
- Division of Biotechnology, CAS Key Laboratory of SSACDalian Institute of Chemical Physics, Chinese Academy of SciencesDalianChina
- University of Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and RegenerationBeijingChina
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of SciencesShanghaiChina
| |
Collapse
|
91
|
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
|
92
|
Mice Placental ECM Components May Provide A Three-Dimensional Placental Microenvironment. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 10:bioengineering10010016. [PMID: 36671588 PMCID: PMC9855196 DOI: 10.3390/bioengineering10010016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/30/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022]
Abstract
Bioethical limitations impair deeper studies in human placental physiology, then most studies use human term placentas or murine models. To overcome these challenges, new models have been proposed to mimetize the placental three-dimensional microenvironment. The placental extracellular matrix plays an essential role in several processes, being a part of the establishment of materno-fetal interaction. Regarding these aspects, this study aimed to investigate term mice placental ECM components, highlighting its collagenous and non-collagenous content, and proposing a potential three-dimensional model to mimetize the placental microenvironment. For that, 18.5-day-old mice placenta, both control and decellularized (n = 3 per group) were analyzed on Orbitrap Fusion Lumos spectrometer (ThermoScientific) and LFQ intensity generated on MaxQuant software. Proteomic analysis identified 2317 proteins. Using ECM and cell junction-related ontologies, 118 (5.1%) proteins were filtered. Control and decellularized conditions had no significant differential expression on 76 (64.4%) ECM and cell junction-related proteins. Enriched ontologies in the cellular component domain were related to cell junction, collagen and lipoprotein particles, biological process domain, cell adhesion, vasculature, proteolysis, ECM organization, and molecular function. Enriched pathways were clustered in cell adhesion and invasion, and labyrinthine vasculature regulation. These preserved ECM proteins are responsible for tissue stiffness and could support cell anchoring, modeling a three-dimensional structure that may allow placental microenvironment reconstruction.
Collapse
|
93
|
The multifaceted role of GCM1 during trophoblast differentiation in the human placenta. Proc Natl Acad Sci U S A 2022; 119:e2203071119. [PMID: 36442132 PMCID: PMC9894182 DOI: 10.1073/pnas.2203071119] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Remodeling of the uterine vasculature by invasive extravillous trophoblasts (EVTs) is a critical aspect of human placentation. Insufficient EVT invasion can lead to severe obstetrical complications like preeclampsia, intrauterine growth restriction, and preterm birth. Glial cells missing-1 (GCM1) is a transcription factor that is crucial for proper placentation in mice, and is highly expressed in human syncytiotrophoblast (ST) and EVTs. GCM1 is classically considered a master regulator of ST formation, but little is known about its contribution to the development and function of EVTs. Therefore, in this study we test the hypothesis that GCM1 is a critical regulator of both EVT and ST development and function. We show that GCM1 is highly expressed in human trophoblast stem (TS) cells differentiated into either ST or EVTs. Knockdown of GCM1 in TS cells hindered differentiation into both ST and EVT pathways. When placed in ST media, GCM1-knockdown cells formed small, unstable clusters; when placed in EVT media, cells had altered morphology and transcript profiles resembling cells trapped in an intermediate state between CT and EVT, and invasive capacity through matrix was reduced. RNA sequencing analysis of GCM1-deficient TS cells revealed downregulation of EVT-associated genes and enrichment in transcripts related to WNT signaling, which was linked to decreased expression of the EVT master regulator ASCL2 and the WNT antagonist NOTUM. Our findings reveal an essential role of GCM1 during ST and EVT development, and suggest that GCM1 regulates differentiation of human TS cells into EVTs by inducing expression of ASCL2 and NOTUM.
Collapse
|
94
|
Transcriptomic mapping of the metzincin landscape in human trophoblasts. Gene Expr Patterns 2022; 46:119283. [PMID: 36307023 DOI: 10.1016/j.gep.2022.119283] [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: 08/08/2022] [Revised: 10/12/2022] [Accepted: 10/17/2022] [Indexed: 11/04/2022]
Abstract
The metzincin family of metalloproteases coordinates tissue developmental processes through regulation of growth factor availability, receptor signaling, and cell-cell/cell-matrix adhesion. While roles for select metzincins in controlling trophoblast functions in human placental development have been described, a comprehensive understanding of metzincin dynamics during trophoblast differentiation is lacking. To address this knowledge gap, single cell transcriptomic datasets derived from first trimester chorionic villi and decidua were used to decipher metzincin expression profiles and kinetics in diverse cell types within the utero-placental interface. Further, specific protease-substrate interactions within progenitor trophoblasts were examined to better define the progenitor niche. Within the uterine-placental compartment, 43 metzincin proteases were expressed across 15 cell-type clusters. Metzincin subgroups expressed in placental trophoblasts, placental mesenchymal cells, uterine stromal, and immune cells included multiple matrix metalloproteases (MMPs), a disintegrin and metalloproteases (ADAMs), a disintegrin and metalloproteases with thrombospondin repeats (ADAMTSs), pappalysins, and astacins. Within the trophoblast compartment, eight distinct trophoblasts states were identified: four cytotrophoblast (CTB), one syncytiotrophoblast precursor (SCTp), two column CTB (cCTB), and one extravillous trophoblast (EVT). Within these states 7 MMP, 8 ADAM, 4 ADAMTS, 2 pappalysin, and 3 astacin proteases were expressed. Cell trajectory modeling shows that expression of most (19/24) metzincins increase during EVT differentiation, though expression of select metalloproteases increase along the villous pathway. Eleven metzincins (ADAM10, -17, MMP14, -15, -19, -23B, ADAMTS1, -6, -19, TLL-1, -2) showed enrichment within CTB progenitors, and analysis of metzincin-substrate interactions identified ∼150 substrates and binding partners, including FBN2 as an ADAMTS6-specific substrate. Together, this work characterizes the metzincin landscape in human first trimester trophoblasts and establishes insight into the roles specific proteases perform within distinct trophoblast niches and across trophoblast differentiation. This resource serves as a guide for future investigations into the roles of metzincin proteases in human placental development.
Collapse
|
95
|
Human Maternal-Fetal Interface Cellular Models to Assess Antiviral Drug Toxicity during Pregnancy. REPRODUCTIVE MEDICINE 2022. [DOI: 10.3390/reprodmed3040024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Pregnancy is a period of elevated risk for viral disease severity, resulting in serious health consequences for both the mother and the fetus; yet antiviral drugs lack comprehensive safety and efficacy data for use among pregnant women. In fact, pregnant women are systematically excluded from therapeutic clinical trials to prevent potential fetal harm. Current FDA-recommended reproductive toxicity assessments are studied using small animals which often do not accurately predict the human toxicological profiles of drug candidates. Here, we review the potential of human maternal-fetal interface cellular models in reproductive toxicity assessment of antiviral drugs. We specifically focus on the 2- and 3-dimensional maternal placental models of different gestational stages and those of fetal embryogenesis and organ development. Screening of drug candidates in physiologically relevant human maternal-fetal cellular models will be beneficial to prioritize selection of safe antiviral therapeutics for clinical trials in pregnant women.
Collapse
|
96
|
Tan JP, Liu X, Polo JM. Establishment of human induced trophoblast stem cells via reprogramming of fibroblasts. Nat Protoc 2022; 17:2739-2759. [PMID: 36241724 DOI: 10.1038/s41596-022-00742-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 06/22/2022] [Indexed: 11/09/2022]
Abstract
During early mammalian embryonic development, trophoblast cells play an essential role in establishing cell-cell interactions at the maternal-fetal interface to ensure a successful pregnancy. In a recent study, we showed that human fibroblasts can be reprogrammed into induced trophoblast stem (iTS) cells by transcription factor-mediated nuclear reprogramming using the Yamanaka factors OCT4, KLF4, SOX2 and c-MYC (OKSM) and a selection of TS cell culture conditions. The derivation of TS cells from human blastocysts or first-trimester placenta can be limited by difficulties in obtaining adequate material as well as ethical implications. By contrast, the described approach allows the generation of iTS cells from the adult cells of individuals with diverse genetic backgrounds, which are readily accessible to many laboratories around the world. Here we describe a step-by-step protocol for the generation and establishment of human iTS cells directly from dermal fibroblasts using a non-integrative reprogramming method. The protocol consists of four main sections: (1) recovery of cryopreserved human dermal fibroblasts, (2) somatic cell reprogramming, (3) passaging of reprogramming intermediates and (4) derivation of iTS cell cultures followed by routine maintenance of iTS cells. These iTS cell lines can be established in 2-3 weeks and cultured long term over 50 passages. We also discuss several characterization methods that can be performed to validate the iTS cells derived using this approach. Our protocol allows researchers to generate patient-specific iTS cells to interrogate the trophoblast and placenta biology as well as their interactions with embryonic cells in health and diseases.
Collapse
Affiliation(s)
- Jia Ping Tan
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Xiaodong Liu
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia.
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia.
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
- School of Life Sciences, Westlake University, Hangzhou, China.
- Westlake Institute for Advanced Study, Hangzhou, China.
| | - Jose M Polo
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia.
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia.
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.
- Adelaide Centre for Epigenetics, Faculty of Medicine Nursing and Medical Sciences, The University of Adelaide, Adelaide, Australia.
- The South Australian Immunogenomics Cancer Institute, Faculty of Medicine Nursing and Medical Sciences, The University of Adelaide, Adelaide, Australia.
| |
Collapse
|
97
|
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
|
98
|
Ruan D, Ye ZW, Yuan S, Li Z, Zhang W, Ong CP, Tang K, Ka Ki Tam TT, Guo J, Xuan Y, Huang Y, Zhang Q, Lee CL, Lu L, Chiu PCN, Yeung WSB, Liu F, Jin DY, Liu P. Human early syncytiotrophoblasts are highly susceptible to SARS-CoV-2 infection. Cell Rep Med 2022; 3:100849. [PMID: 36495872 PMCID: PMC9671691 DOI: 10.1016/j.xcrm.2022.100849] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 08/23/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022]
Abstract
Direct in vivo investigation of human placenta trophoblast's susceptibility to SARS-CoV-2 is challenging. Here we report that human trophoblast stem cells (hTSCs) and their derivatives are susceptible to SARS-CoV-2 infection, which reveals heterogeneity in hTSC cultures. Early syncytiotrophoblasts (eSTBs) generated from hTSCs have enriched transcriptomic features of peri-implantation trophoblasts, express high levels of angiotensin-converting enzyme 2 (ACE2), and are productively infected by SARS-CoV-2 and its Delta and Omicron variants to produce virions. Antiviral drugs suppress SARS-CoV-2 replication in eSTBs and antagonize the virus-induced blockage of STB maturation. Although less susceptible to SARS-CoV-2 infection, trophoblast organoids originating from hTSCs show detectable viral replication reminiscent of the uncommon placental infection. These findings implicate possible risk of COVID-19 infection in peri-implantation embryos, which may go unnoticed. Stem cell-derived human trophoblasts such as eSTBs can potentially provide unlimited amounts of normal and genome-edited cells and facilitate coronavirus research and antiviral discovery.
Collapse
Affiliation(s)
- Degong Ruan
- Centre for Translational Stem Cell Biology, The University of Hong Kong, Hong Kong Special Administrative Region, China; Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Zi-Wei Ye
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Shuofeng Yuan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Zhuoxuan Li
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Weiyu Zhang
- Centre for Translational Stem Cell Biology, The University of Hong Kong, Hong Kong Special Administrative Region, China; Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Chon Phin Ong
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Kaiming Tang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Timothy Theodore Ka Ki Tam
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jilong Guo
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yiyi Xuan
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yunying Huang
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Qingqing Zhang
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Cheuk-Lun Lee
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Liming Lu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Philip C N Chiu
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - William S B Yeung
- Centre for Translational Stem Cell Biology, The University of Hong Kong, Hong Kong Special Administrative Region, China; Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China; Department of Obstetrics and Gynaecology, School of Clinical Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Fang Liu
- Centre for Translational Stem Cell Biology, The University of Hong Kong, Hong Kong Special Administrative Region, China; Foshan Stomatology Hospital, School of Medicine, Foshan University, No. 5 Hebing Road, Foshan, Guangdong Province, China.
| | - Dong-Yan Jin
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| | - Pentao Liu
- Centre for Translational Stem Cell Biology, The University of Hong Kong, Hong Kong Special Administrative Region, China; Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China; Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China; School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| |
Collapse
|
99
|
Cox BJ, Naismith K. Here and there a trophoblast, a transcriptional evaluation of trophoblast cell models. Cell Mol Life Sci 2022; 79:584. [PMID: 36346530 DOI: 10.1007/s00018-022-04589-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 09/23/2022] [Accepted: 10/06/2022] [Indexed: 11/10/2022]
Abstract
A recent explosion of methods to produce human trophoblast and stem cells (hTSCs) is fuelling a renewed interest in this tissue. The trophoblast is critical to reproduction by facilitating implantation, maternal physiological adaptations to pregnancy and the growth of the fetus through transport of nutrients between the mother and fetus. More broadly, the trophoblast has phenotypic properties that make it of interest to other fields. Its angiogenic and invasive properties are similar to tumours and could identify novel drug targets, and its ability to regulate immunological tolerance of the allogenic fetus could lead to improvements in transplantations. Within this review, we integrate and assess transcriptomic data of cell-based models of hTSC alongside in vivo samples to identify the utility and applicability of these models. We also integrate single-cell RNA sequencing data sets of human blastoids, stem cells and embryos to identify how these models may recapitulate early trophoblast development.
Collapse
Affiliation(s)
- Brian J Cox
- Department of Physiology, University of Toronto, 1 King's College Circle, MS 3360, Toronto, ON, M6J2J2, Canada. .,Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON, Canada.
| | - Kendra Naismith
- Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| |
Collapse
|
100
|
Cindrova-Davies T, Sferruzzi-Perri AN. Human placental development and function. Semin Cell Dev Biol 2022; 131:66-77. [PMID: 35393235 DOI: 10.1016/j.semcdb.2022.03.039] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/30/2022] [Accepted: 03/30/2022] [Indexed: 12/14/2022]
Abstract
The placenta is a transient fetal organ that plays a critical role in the health and wellbeing of both the fetus and its mother. Functionally, the placenta sustains the growth of the fetus as it facilitates delivery of oxygen and nutrients and removal of waste products. Not surprisingly, defective early placental development is the primary cause of common disorders of pregnancy, including recurrent miscarriage, fetal growth restriction, pre-eclampsia and stillbirth. Adverse pregnancy conditions will also affect the life-long health of the fetus via developmental programming[1]. Despite its critical importance in reproductive success and life-long health, our understanding of placental development is not extensive, largely due to ethical limitations to studying early or chronological placental development, lack of long-term in vitro models, or comparative animal models. In this review, we examine current knowledge of early human placental development, discuss the critical role of the maternal endometrium and of the fetal-maternal dialogue in pregnancy success, and we explore the latest models of trophoblast and endometrial stem cells. In addition, we discuss the role of oxygen in placental formation and function, how nutrient delivery is mediated during the periods of histotrophic nutrition (uptake of uterine secretions) and haemotrophic nutrition (exchange between the maternal and fetal circulations), and how placental endocrine function facilitates fetal growth and development.
Collapse
Affiliation(s)
- Tereza Cindrova-Davies
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Amanda N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK.
| |
Collapse
|