1
|
Keding LT, Heselton AR, Ren E, Shaw SA, Koenig MR, Golos TG, Schmidt JK. In vitro differentiation of macaque extravillous trophoblasts in a low oxygen environment. Placenta 2025; 163:16-28. [PMID: 40024139 PMCID: PMC11955297 DOI: 10.1016/j.placenta.2025.02.014] [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: 07/23/2024] [Revised: 02/07/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
INTRODUCTION Early primate placental development occurs within a low oxygen (O2) environment, whereas in vitro differentiation of trophoblasts is performed at supraphysiologic O2 levels. Macaque trophoblast stem cells (TSCs) are capable of differentiation to extravillous trophoblasts (EVTs) in vitro, yet the morphological heterogeneity observed across cells lines necessitates evaluation of optimal culture conditions. Our objectives were to determine the impact of oxygen on the in vitro differentiation of macaque TSCs and to refine the molecular characterization of TSC-differentiated EVTs. METHODS Macaque TSCs were differentiated to EVTs in either 20% or 5% O2. Gene and protein expression profiles were compared between TSCs and EVTs and between differentiation conditions. Immunohistochemical analysis was performed on early gestation macaque placental tissues to assess in vivo expression of Ki-67, NCAM1 and monkey chorionic gonadotropin (mCG). RESULTS EVTs differentiated in 20% O2 had significantly higher expression of CGA, CGB and NOTCH2 and decreased HIF1A expression compared to those cultured in 5% O2. Regardless of oxygen condition, nearly all EVTs expressed NCAM1 and Mamu-AG, the macaque-specific homolog of human EVT marker HLA-G. In vivo placental expression of NCAM1 was restricted to EVTs within the trophoblastic shell and endovasculature, revealing a macaque EVT marker within the placenta. DISCUSSION Reduced oxygen minimally impacted macaque EVT differentiation in vitro. Elevated expression of the endovascular EVT marker NOTCH2 potentially suggests that 20% O2 supported differentiation of more mature EVTs. Altogether, a standard 20% O2 environment supports macaque EVT differentiation in vitro and the results further validate the identity of macaque TSC-differentiated EVTs.
Collapse
Affiliation(s)
- Logan T Keding
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Ct, Madison, WI, 53715, USA; Department of Obstetrics and Gynecology, University of Wisconsin-Madison, 202 South Park St, Madison, WI, 53715, USA
| | - Avery R Heselton
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Ct, Madison, WI, 53715, USA
| | - Emily Ren
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Ct, Madison, WI, 53715, USA
| | - Sarah A Shaw
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Ct, Madison, WI, 53715, USA
| | - Michelle R Koenig
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Ct, Madison, WI, 53715, USA
| | - Thaddeus G Golos
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Ct, Madison, WI, 53715, USA; Department of Obstetrics and Gynecology, University of Wisconsin-Madison, 202 South Park St, Madison, WI, 53715, USA; Department of Comparative Biosciences, School of Veterinary Medicine, 2015 Linden Dr, Madison, WI, 53706, USA
| | - Jenna K Schmidt
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Ct, Madison, WI, 53715, USA.
| |
Collapse
|
2
|
Dimova T, Alexandrova M, Vangelov I, You Y, Mor G. The modeling of human implantation and early placentation: achievements and perspectives. Hum Reprod Update 2025; 31:133-163. [PMID: 39673726 DOI: 10.1093/humupd/dmae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 10/29/2024] [Indexed: 12/16/2024] Open
Abstract
BACKGROUND Successful implantation is a critical step for embryo survival. The major losses in natural and assisted human reproduction appeared to occur during the peri-implantation period. Because of ethical constraints, the fascinating maternal-fetal crosstalk during human implantation is difficult to study and thus, the possibility for clinical intervention is still limited. OBJECTIVE AND RATIONALE This review highlights some features of human implantation as a unique, ineffective and difficult-to-model process and summarizes the pros and cons of the most used in vivo, ex vivo and in vitro models. We point out the variety of cell line-derived models and how these data are corroborated by well-defined primary cells of the same nature. Important aspects related to the handling, standardization, validation, and modus operandi of the advanced 3D in vitro models are widely discussed. Special attention is paid to blastocyst-like models recapitulating the hybrid phenotype and HLA profile of extravillous trophoblasts, which are a unique yet poorly understood population with a major role in the successful implantation and immune mother-embryo recognition. Despite raising new ethical dilemmas, extended embryo cultures and synthetic embryo models are also in the scope of our review. SEARCH METHODS We searched the electronic database PubMed from inception until March 2024 by using a multi-stage search strategy of MeSH terms and keywords. In addition, we conducted a forward and backward reference search of authors mentioned in selected articles. OUTCOMES Primates and rodents are valuable in vivo models for human implantation research. However, the deep interstitial, glandular, and endovascular invasion accompanied by a range of human-specific factors responsible for the survival of the fetus determines the uniqueness of the human implantation and limits the cross-species extrapolation of the data. The ex vivo models are short-term cultures, not relevant to the period of implantation, and difficult to standardize. Moreover, the access to tissues from elective terminations of pregnancy raises ethical and legal concerns. Easy-to-culture cancer cell lines have many limitations such as being prone to spontaneous transformation and lacking decent tissue characteristics. The replacement of the original human explants, primary cells or cancer cell lines with cultures of immortalized cell lines with preserved stem cell characteristics appears to be superior for in vitro modeling of human implantation and early placentation. Remarkable advances in our understanding of the peri-implantation stages have also been made by advanced three dimensional (3D) models i.e. spheroids, organoids, and assembloids, as placental and endometrial surrogates. Much work remains to be done for the optimization and standardization of these integrated and complex models. The inclusion of immune components in these models would be an asset to delineate mechanisms of immune tolerance. Stem cell-based embryo-like models and surplus IVF embryos for research bring intriguing possibilities and are thought to be the trend for the next decade for in vitro modeling of human implantation and early embryogenesis. Along with this research, new ethical dilemmas such as the moral status of the human embryo and the potential exploitation of women consenting to donate their spare embryos have emerged. The careful appraisal and development of national legal and ethical frameworks are crucial for better regulation of studies using human embryos and embryoids to reach the potential benefits for human reproduction. WIDER IMPLICATIONS We believe that our data provide a systematization of the available information on the modeling of human implantation and early placentation and will facilitate further research in this field. A strict classification of the advanced 3D models with their pros, cons, applicability, and availability would help improve the research quality to provide reliable outputs.
Collapse
Affiliation(s)
- Tanya Dimova
- Institute of Biology and Immunology of Reproduction "Acad. Kiril Bratanov", Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Marina Alexandrova
- Institute of Biology and Immunology of Reproduction "Acad. Kiril Bratanov", Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Ivaylo Vangelov
- Institute of Biology and Immunology of Reproduction "Acad. Kiril Bratanov", Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Yuan You
- C.S. Mott Center for Human Growth and Development, Wayne State University, Detroit, MI, USA
| | - Gil Mor
- C.S. Mott Center for Human Growth and Development, Wayne State University, Detroit, MI, USA
| |
Collapse
|
3
|
van Voorden AJ, Boussata S, Keijser R, Vermij M, Wagner MK, Ganzevoort W, Afink GB. Generation of Bona Fide Human Induced Trophoblast Stem Cells by Direct Reprogramming of Term Umbilical Cord Cells. Int J Mol Sci 2024; 26:271. [PMID: 39796127 PMCID: PMC11719488 DOI: 10.3390/ijms26010271] [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: 11/28/2024] [Revised: 12/23/2024] [Accepted: 12/25/2024] [Indexed: 01/13/2025] Open
Abstract
Placentation disorders, including severe preeclampsia and fetal growth restriction, have their origins in early pregnancy, whereas symptoms typically present later on. To investigate the pathogenesis of these diseases, there is a need for a reliable in vitro model system of early placenta development with known pregnancy outcomes. Therefore, we optimized the generation of human induced trophoblast stem cells (iTSCs) from term umbilical cord, enabling non-invasive collection of patient-derived material immediately after birth. Using a direct reprogramming approach previously described for dermal fibroblasts, we investigated the effects of three supplements (A-485, BMP4, and EPZ-6438) to assess their potential to enhance iTSC induction. The generated iTSCs fulfilled the criteria for bona fide first-trimester trophoblasts and exhibited key functional capacities, including long-term self-renewal, differentiation into hormone-producing syncytiotrophoblasts and invasive extravillous trophoblasts, and the formation of organoids. Furthermore, transcriptomic analysis revealed high similarity between the generated iTSCs and trophoblast stem cells derived from first-trimester placental tissue. The supplements did not improve the generation of iTSCs. In conclusion, we successfully generated bona fide iTSCs from term umbilical cord using a direct reprogramming approach, providing a robust and clinically relevant model to study early placentation mechanisms in patient-derived trophoblasts.
Collapse
Affiliation(s)
- A. Jantine van Voorden
- Reproductive Biology Laboratory, Amsterdam University Medical Center Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, 1105 AZ Amsterdam, The Netherlands
| | - Souad Boussata
- Reproductive Biology Laboratory, Amsterdam University Medical Center Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, 1105 AZ Amsterdam, The Netherlands
| | - Remco Keijser
- Reproductive Biology Laboratory, Amsterdam University Medical Center Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, 1105 AZ Amsterdam, The Netherlands
| | - Marloes Vermij
- Reproductive Biology Laboratory, Amsterdam University Medical Center Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, 1105 AZ Amsterdam, The Netherlands
| | - Muriel K. Wagner
- Reproductive Biology Laboratory, Amsterdam University Medical Center Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, 1105 AZ Amsterdam, The Netherlands
| | - Wessel Ganzevoort
- Amsterdam Reproduction and Development Research Institute, 1105 AZ Amsterdam, The Netherlands
- Department of Obstetrics and Gynaecology, Amsterdam University Medical Center Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Gijs B. Afink
- Reproductive Biology Laboratory, Amsterdam University Medical Center Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, 1105 AZ Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
4
|
Rong L, Xiang L, Ai Z, Niu B, Wang Y, Yin Y, Feng C, Shi G, Chen T, Yang J, Luo X, Bai Y, Zhou X, Liu X, Zheng H, Ke Y, Li T, Wu Z. The impact of dynamic caudal type homeobox 2 expression on the differentiation of human trophoblast lineage during implantation. Cell Prolif 2024; 57:e13729. [PMID: 39161168 PMCID: PMC11628739 DOI: 10.1111/cpr.13729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/21/2024] [Accepted: 07/23/2024] [Indexed: 08/21/2024] Open
Abstract
The trophoblast lineage differentiation represents a rate-limiting step in successful embryo implantation. Adhesion, invasion and migration processes within the trophoblast are governed by several transcription factors. Among them, CDX2 is a critical regulator shaping the destiny of the trophoblast. While its altered expression is a linchpin initiating embryo implantation in mice, the precise influence of CDX2 on the functionality and lineage differentiation of early human trophoblast remains unclear. In this study, we employed well-established human trophoblast stem cell (hTSC) lines with CDX2 overexpression coupled with a 3D in vitro culture system for early human embryos. We revealed that the downregulation of CDX2 is a prerequisite for syncytialization during human embryo implantation based on immunofluorescence, transcriptome analysis, CUT-tag sequencing and the construction of 3D human trophoblast organoids. While CDX2 overexpression inhibited syncytialization, it propelled hTSC proliferation and invasive migration. CDX2 exerted its influence by interacting with CGA, PTGS2, GCM1, LEF1 and CDH2, thereby hindering premature differentiation of the syncytiotrophoblast. CDX2 overexpression enhanced the epithelial-mesenchymal transition of human trophoblast organoids. In summary, our study provides insights into the molecular characteristics of trophoblast differentiation and development in humans, laying a theoretical foundation for advancing research in embryo implantation.
Collapse
Affiliation(s)
- Lujuan Rong
- Faculty of Life Science and TechnologyKunming University of Science and TechnologyKunmingYunnanChina
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- The Affiliated Hospital of Kunming University of Science and TechnologyKunmingYunnanChina
- Department of Reproductive Medicine, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China (Co‐building)The First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnanChina
| | - Lifeng Xiang
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- The Affiliated Hospital of Kunming University of Science and TechnologyKunmingYunnanChina
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnanChina
- KUST‐YPFPH Reproductive Medicine Joint Research CenterKunmingYunnanChina
| | - Zongyong Ai
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnanChina
| | - Baohua Niu
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnanChina
| | - Yaqing Wang
- University of Science and Technology of ChinaHefeiAnhuiChina
- Suzhou Institute for Advanced ResearchUniversity of Science and Technology of ChinaSuzhouJiangsuChina
| | - Yu Yin
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnanChina
| | - Chun Feng
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnanChina
| | - Gaohui Shi
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnanChina
| | - Tingwei Chen
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnanChina
| | - Jie Yang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnanChina
| | - Xi Luo
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- The Affiliated Hospital of Kunming University of Science and TechnologyKunmingYunnanChina
- Department of Reproductive Medicine, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China (Co‐building)The First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- KUST‐YPFPH Reproductive Medicine Joint Research CenterKunmingYunnanChina
| | - Yun Bai
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- The Affiliated Hospital of Kunming University of Science and TechnologyKunmingYunnanChina
- Department of Reproductive Medicine, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China (Co‐building)The First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- KUST‐YPFPH Reproductive Medicine Joint Research CenterKunmingYunnanChina
| | - Xiaoting Zhou
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- The Affiliated Hospital of Kunming University of Science and TechnologyKunmingYunnanChina
- Department of Reproductive Medicine, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China (Co‐building)The First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- KUST‐YPFPH Reproductive Medicine Joint Research CenterKunmingYunnanChina
| | - Xiaoping Liu
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- The Affiliated Hospital of Kunming University of Science and TechnologyKunmingYunnanChina
- Department of Reproductive Medicine, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China (Co‐building)The First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- KUST‐YPFPH Reproductive Medicine Joint Research CenterKunmingYunnanChina
| | - Haishan Zheng
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- The Affiliated Hospital of Kunming University of Science and TechnologyKunmingYunnanChina
- Department of Reproductive Medicine, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China (Co‐building)The First People's Hospital of Yunnan ProvinceKunmingYunnanChina
| | - Yang Ke
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- The Affiliated Hospital of Kunming University of Science and TechnologyKunmingYunnanChina
- Department of Reproductive Medicine, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China (Co‐building)The First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- KUST‐YPFPH Reproductive Medicine Joint Research CenterKunmingYunnanChina
| | - Tianqing Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnanChina
| | - Ze Wu
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- The Affiliated Hospital of Kunming University of Science and TechnologyKunmingYunnanChina
- Department of Reproductive Medicine, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China (Co‐building)The First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- KUST‐YPFPH Reproductive Medicine Joint Research CenterKunmingYunnanChina
| |
Collapse
|
5
|
Lai Y, Fu Z, Gao Y, Ma N, Li L. Hypoxia-inducible factors (HIFs) in early pregnancy: implications for miscarriage†. Biol Reprod 2024; 111:987-999. [PMID: 39325972 DOI: 10.1093/biolre/ioae139] [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: 05/09/2024] [Revised: 09/07/2024] [Accepted: 09/25/2024] [Indexed: 09/28/2024] Open
Abstract
Miscarriage poses a significant threat to both maternal and fetal health. Its etiology remains unknown, and there are no established effective identification or prevention strategies. A low-oxygen environment in early pregnancy is a physiological necessity for embryonic and placental growth. Hypoxia-inducible factors are a family of classic hypoxia signaling molecules whose expression level may fluctuate abnormally because of an imbalance in oxygen levels. Its unusual fluctuations initiate multiple signaling pathways at the maternal womb. Hypoxia-inducible factors are a family of classic hypoxia-signaling molecules and immune tolerance. Notably, aberrant regulation of these processes may lead to miscarriage. This review aims to clarify how the hypoxia-inducible factor-1α mediates the aberrant regulation of biological processes, including autophagy, metabolic reprogramming, et al., and how these effects impact trophoblasts and other cells at the maternal-fetal interface. These findings provide new insights into potential therapeutic and preventive strategies for miscarriage.
Collapse
Affiliation(s)
- Yuxuan Lai
- Department of Social Medicine and Health Care Management, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Zhiyu Fu
- Department of Social Medicine and Health Care Management, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Yaxin Gao
- Department of Social Medicine and Health Care Management, School of Public Health, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Environment and Population Health, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Ning Ma
- Department of Social Medicine and Health Care Management, School of Public Health, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Environment and Population Health, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Lu Li
- Department of Social Medicine and Health Care Management, School of Public Health, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Environment and Population Health, School of Public Health, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
6
|
Azamor T, Familiar-Macedo D, Salem GM, Onwubueke C, Melano I, Bian L, Vasconcelos Z, Nielsen-Saines K, Wu X, Jung JU, Lin F, Goje O, Chien E, Gordon S, Foster CB, Aly H, Farrell RM, Chen W, Foo SS. Transplacental SARS-CoV-2 protein ORF8 binds to complement C1q to trigger fetal inflammation. EMBO J 2024; 43:5494-5529. [PMID: 39390219 PMCID: PMC11574245 DOI: 10.1038/s44318-024-00260-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 09/08/2024] [Accepted: 09/12/2024] [Indexed: 10/12/2024] Open
Abstract
Prenatal SARS-CoV-2 infection is associated with higher rates of pregnancy and birth complications, despite that vertical transmission rates are thought to be low. Here, multi-omics analyses of human placental tissues, cord tissues/plasma, and amniotic fluid from 23 COVID-19 mother-infant pairs revealed robust inflammatory responses in both maternal and fetal compartments. Pronounced expression of complement proteins (C1q, C3, C3b, C4, C5) and inflammatory cytokines (TNF, IL-1α, and IL-17A/E) was detected in the fetal compartment of COVID-19-affected pregnancies. While ~26% of fetal tissues were positive for SARS-CoV-2 RNA, more than 60% of fetal tissues contained SARS-CoV-2 ORF8 proteins, suggesting transplacental transfer of this viral accessory protein. ORF8-positive fetal compartments exhibited increased inflammation and complement activation compared to ORF8-negative COVID-19 pregnancies. In human placental trophoblasts in vitro, exogenous ORF8 exposure resulted in complement activation and inflammatory responses. Co-immunoprecipitation analysis demonstrated that ORF8 binds to C1q specifically by interacting with a 15-peptide region on ORF8 (C37-A51) and the globular domain of C1q subunit A. In conclusion, an ORF8-C1q-dependent complement activation pathway was identified in COVID-19-affected pregnancies, likely contributing to fetal inflammation independently of fetal virus exposure.
Collapse
Grants
- R00DE028573 HHS | NIH | National Institute of Dental and Craniofacial Research (NIDR)
- R01 AI116585 NIAID NIH HHS
- R01AI140705,R01AI140718,R01AI116585,AI140718,AI172252 HHS | National Institutes of Health (NIH)
- AI129534,AI298847,AI140718,AI172252,AI140718,AI172252 HHS | National Institutes of Health (NIH)
- R01 AI140718 NIAID NIH HHS
- R01 DE033391 NIDCR NIH HHS
- RES515531 Clinical and Translational Science Center, Weill Cornell Medicine (CTSC)
- R21 AI129534 NIAID NIH HHS
- R01 AI140705 NIAID NIH HHS
- R00 DE028573 NIDCR NIH HHS
- R01DE033391 HHS | NIH | National Institute of Dental and Craniofacial Research (NIDR)
- S10 OD023436 NIH HHS
- R56 AI172252 NIAID NIH HHS
- UM1 TR004528 NCATS NIH HHS
Collapse
Affiliation(s)
- Tamiris Azamor
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Débora Familiar-Macedo
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Gielenny M Salem
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Chineme Onwubueke
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Ivonne Melano
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Lu Bian
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - Karin Nielsen-Saines
- Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Xianfang Wu
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Jae U Jung
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Feng Lin
- Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Oluwatosin Goje
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Edward Chien
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Steve Gordon
- Section of Pediatric Infectious Diseases, Children's Institute, Cleveland Clinic Children's, Cleveland Clinic, Cleveland, OH, USA
| | - Charles B Foster
- Section of Pediatric Infectious Diseases, Children's Institute, Cleveland Clinic Children's, Cleveland Clinic, Cleveland, OH, USA
| | - Hany Aly
- Cleveland Clinic Children's, Cleveland Clinic, Cleveland, OH, USA
| | - Ruth M Farrell
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Institute, Cleveland Clinic, Cleveland, OH, USA.
| | - Weiqiang Chen
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA.
| | - Suan-Sin Foo
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
7
|
Slamecka J, Ryu S, Tristan CA, Chu PH, Weber C, Deng T, Gedik Y, Ormanoglu P, Voss TC, Simeonov A, Singeç I. Highly efficient generation of self-renewing trophoblast from human pluripotent stem cells. iScience 2024; 27:110874. [PMID: 39386760 PMCID: PMC11462042 DOI: 10.1016/j.isci.2024.110874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 06/02/2024] [Accepted: 08/30/2024] [Indexed: 10/12/2024] Open
Abstract
Human pluripotent stem cells (hPSCs) represent a powerful model system to study early developmental processes. However, lineage specification into trophectoderm (TE) and trophoblast (TB) differentiation remains poorly understood, and access to well-characterized placental cells for biomedical research is limited, largely depending on fetal tissues or cancer cell lines. Here, we developed novel strategies enabling highly efficient TE specification that generates cytotrophoblast (CTB) and multinucleated syncytiotrophoblast (STB), followed by the establishment of trophoblast stem cells (TSCs) capable of differentiating into extravillous trophoblast (EVT) and STB after long-term expansion. We confirmed stepwise and controlled induction of lineage- and cell-type-specific genes consistent with developmental biology principles and benchmarked typical features of placental cells using morphological, biochemical, genomics, epigenomics, and single-cell analyses. Charting a well-defined roadmap from hPSCs to distinct placental phenotypes provides invaluable opportunities for studying early human development, infertility, and pregnancy-associated diseases.
Collapse
Affiliation(s)
- Jaroslav Slamecka
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Seungmi Ryu
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Carlos A. Tristan
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Pei-Hsuan Chu
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Claire Weber
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Tao Deng
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Yeliz Gedik
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Pinar Ormanoglu
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Ty C. Voss
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Anton Simeonov
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Ilyas Singeç
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| |
Collapse
|
8
|
Fu S, Huang B, Li E, Xu X, Xu RH. Chimerization of human ESC-derived extraembryonic cells with the mouse blastocyst. Int J Biol Sci 2024; 20:5056-5069. [PMID: 39430245 PMCID: PMC11488584 DOI: 10.7150/ijbs.99519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/06/2024] [Indexed: 10/22/2024] Open
Abstract
It has been reported that human embryonic stem cells (hESCs) treated with BMP4 and inhibitors of TGFβ signaling (A83-01) and FGF signaling (PD173074), called BAP, can efficiently differentiate to extraembryonic (ExE) cells in vitro. Due to restricted access to human embryos, it is ethically impossible to test the developmental potential of ExE cells in vivo. Here, we demonstrate that most ExE cells expressed molecular markers for both trophoblasts (TBs) and amniotic cells (ACs). Following intra-uterine transplantation, ExE cells contributed to the mouse placenta. More interestingly, ExE cells could chimerize with the mouse blastocyst as, after injection into the blastocyst, they penetrated its trophectoderm. After implantation of the injected blastocysts into surrogate mice, human cells were found at E14 in placental labyrinth, junction zones, and even near the uterine decidua, expressed placental markers, and secreted human chorionic gonadotropin. Surprisingly, ExE cells also contributed to cartilages of the chimeric embryo with some expressing the chondrogenic marker SOX9, consistent with the mesodermal potential of TBs and ACs in the placenta. Deleting MSX2, a mesodermal determinant, restricted the contribution of ExE cells to the placenta. Thus, we conclude that hESC-derived ExE cells can chimerize with the mouse blastocyst and contribute to both the placenta and cartilages of the chimera consistent with their heteogenious nature. Intra-uterus and intra-blastocyst injections are novel and sensitive methods to study the developmental potential of ExE cells.
Collapse
Affiliation(s)
| | | | | | | | - Ren-He Xu
- Center of Reproduction, Development & Aging, and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| |
Collapse
|
9
|
Lemke KA, Sarkar CA, Azarin SM. Rapid retinoic acid-induced trophoblast cell model from human induced pluripotent stem cells. Sci Rep 2024; 14:18204. [PMID: 39107470 PMCID: PMC11303561 DOI: 10.1038/s41598-024-68952-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
A limited number of accessible and representative models of human trophoblast cells currently exist for the study of placentation. Current stem cell models involve either a transition through a naïve stem cell state or precise dynamic control of multiple growth factors and small-molecule cues. Here, we demonstrated that a simple five-day treatment of human induced pluripotent stem cells with two small molecules, retinoic acid (RA) and Wnt agonist CHIR 99021 (CHIR), resulted in rapid, synergistic upregulation of CDX2. Transcriptomic analysis of RA + CHIR-treated cells showed high similarity to primary trophectoderm cells. Multipotency was verified via further differentiation towards cells with syncytiotrophoblast or extravillous trophoblast features. RA + CHIR-treated cells were also assessed for the established criteria defining a trophoblast cell model, and they possess all the features necessary to be considered valid. Collectively, our data demonstrate a facile, scalable method for generating functional trophoblast-like cells in vitro to better understand the placenta.
Collapse
Affiliation(s)
- Kristen A Lemke
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Casim A Sarkar
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Samira M Azarin
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
10
|
Motomura K, Morita H, Yamamoto H, Wada S, Sago H, Takahashi H, Saito H, Matsumoto K. Isolation of pure primary term human cytotrophoblasts and their differentiation into syncytiotrophoblast-like cells as an ex vivo model of the human placenta. Placenta 2024:S0143-4004(24)00603-9. [PMID: 39089887 DOI: 10.1016/j.placenta.2024.07.307] [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: 03/29/2024] [Revised: 07/10/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024]
Abstract
The placenta plays a fundamental role in fetal growth and maintenance of pregnancy. Its cellular components include a large multinucleated syncytiotrophoblast (STB) and its progenitor, cytotrophoblasts (CTBs), both of which perform vital functions in the human placenta. Primary cytotrophoblasts isolated from term human placentas that spontaneously fuse and differentiate into syncytiotrophoblast-like cells in vitro have been utilized to investigate the functions of the syncytiotrophoblast and placenta with multiple modifications. Although recent advances have enabled the use of trophoblast stem cell-derived organoids as a model for villous trophoblasts, primary CTBs offer several advantages, including spontaneous differentiation, easy access to materials (from term-delivered human placentas), and simple methodology. Here, we present a precise step-by-step process for isolating pure CTBs from term human placenta based on previously reported placenta digestion, density centrifugation, and CTB purification using anti-HLA-A, B, C antibody. Subsequently, we provide a method to improve CTB viability and differentiation into STB-like cells using epidermal growth factor (EGF) and a ROCK inhibitor (Y-27632) that ensures long-term and stable cultures without altering their proliferation. Because these cells can grow on standard tissue culture plates, this model can be easily utilized for various placental investigations, including innate immune responses, drug resistance, and STB metabolism. Employing this approach considerably enhances our understanding of placental functions, which are key to maternal and offspring health.
Collapse
Affiliation(s)
- Kenichiro Motomura
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan; Center for Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Tokyo, Japan.
| | - Hideaki Morita
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan; Allergy Center, National Center for Child Health and Development, Tokyo, Japan
| | - Hiromichi Yamamoto
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan; Department of Obstetrics and Gynecology, Jichi Medical University, Tochigi, Japan
| | - Seiji Wada
- Center for Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Haruhiko Sago
- Center for Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Tokyo, Japan; Center for Medical Genetics, National Center for Child Health and Development, Tokyo, Japan
| | - Hironori Takahashi
- Department of Obstetrics and Gynecology, Jichi Medical University, Tochigi, Japan
| | - Hirohisa Saito
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Kenji Matsumoto
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan.
| |
Collapse
|
11
|
Bi S, Huang L, Chen Y, Hu Z, Li S, Wang Y, Huang B, Zhang L, Huang Y, Dai B, Du L, Tu Z, Wang Y, Xu D, Xu X, Sun W, Kzhyshkowska J, Wang H, Chen D, Wang F, Zhang S. KAT8-mediated H4K16ac is essential for sustaining trophoblast self-renewal and proliferation via regulating CDX2. Nat Commun 2024; 15:5602. [PMID: 38961108 PMCID: PMC11222414 DOI: 10.1038/s41467-024-49930-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 06/24/2024] [Indexed: 07/05/2024] Open
Abstract
Abnormal trophoblast self-renewal and differentiation during early gestation is the major cause of miscarriage, yet the underlying regulatory mechanisms remain elusive. Here, we show that trophoblast specific deletion of Kat8, a MYST family histone acetyltransferase, leads to extraembryonic ectoderm abnormalities and embryonic lethality. Employing RNA-seq and CUT&Tag analyses on trophoblast stem cells (TSCs), we further discover that KAT8 regulates the transcriptional activation of the trophoblast stemness marker, CDX2, via acetylating H4K16. Remarkably, CDX2 overexpression partially rescues the defects arising from Kat8 knockout. Moreover, increasing H4K16ac via using deacetylase SIRT1 inhibitor, EX527, restores CDX2 levels and promoted placental development. Clinical analysis shows reduced KAT8, CDX2 and H4K16ac expression are associated with recurrent pregnancy loss (RPL). Trophoblast organoids derived from these patients exhibit impaired TSC self-renewal and growth, which are significantly ameliorated with EX527 treatment. These findings suggest the therapeutic potential of targeting the KAT8-H4K16ac-CDX2 axis for mitigating RPL, shedding light on early gestational abnormalities.
Collapse
Affiliation(s)
- Shilei Bi
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Lijun Huang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Yongjie Chen
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Zhenhua Hu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Shanze Li
- National Institute of Biological Sciences, Beijing, 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 102206, China
| | - Yifan Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Baoying Huang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Lizi Zhang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Yuanyuan Huang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Beibei Dai
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Lili Du
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Zhaowei Tu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Yijing Wang
- National Institute of Biological Sciences, Beijing, 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 102206, China
| | - Dan Xu
- National Institute of Biological Sciences, Beijing, 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 102206, China
| | - Xiaotong Xu
- National Institute of Biological Sciences, Beijing, 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 102206, China
| | - Wen Sun
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Julia Kzhyshkowska
- Institute of Transfusion Medicine and Immunology, Mannheim Institute of Innate Immunosciences (MI3), Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany
- German Red Cross Blood Service Baden-Württemberg-Hessen, 68167, Mannheim, Germany
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk, Russia
| | - Haibin Wang
- 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, 361102, China.
| | - Dunjin Chen
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
- Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| | - Fengchao Wang
- National Institute of Biological Sciences, Beijing, 102206, China.
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 102206, China.
| | - Shuang Zhang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
- Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| |
Collapse
|
12
|
Morey R, Bui T, Cheung VC, Dong C, Zemke JE, Requena D, Arora H, Jackson MG, Pizzo D, Theunissen TW, Horii M. iPSC-based modeling of preeclampsia identifies epigenetic defects in extravillous trophoblast differentiation. iScience 2024; 27:109569. [PMID: 38623329 PMCID: PMC11016801 DOI: 10.1016/j.isci.2024.109569] [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: 11/30/2023] [Revised: 02/20/2024] [Accepted: 03/22/2024] [Indexed: 04/17/2024] Open
Abstract
Preeclampsia (PE) is a hypertensive pregnancy disorder with increased risk of maternal and fetal morbidity and mortality. Abnormal extravillous trophoblast (EVT) development and function is considered to be the underlying cause of PE, but has not been previously modeled in vitro. We previously derived induced pluripotent stem cells (iPSCs) from placentas of PE patients and characterized abnormalities in formation of syncytiotrophoblast and responses to changes in oxygen tension. In this study, we converted these primed iPSC to naïve iPSC, and then derived trophoblast stem cells (TSCs) and EVT to evaluate molecular mechanisms underlying PE. We found that primed (but not naïve) iPSC-derived PE-EVT have reduced surface HLA-G, blunted invasive capacity, and altered EVT-specific gene expression. These abnormalities correlated with promoter hypermethylation of genes associated with the epithelial-mesenchymal transition pathway, specifically in primed-iPSC derived PE-EVT. Our findings indicate that abnormal epigenetic regulation might play a role in PE pathogenesis.
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
- Center for Perinatal Discovery, 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
- Center for Perinatal Discovery, University of California San Diego, La Jolla, CA 92093, USA
| | - Virginia Chu Cheung
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Center for Perinatal Discovery, University of California San Diego, La Jolla, CA 92093, USA
| | - Chen Dong
- Department of Developmental Biology and Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joseph E. Zemke
- Department of Developmental Biology and Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Daniela Requena
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Center for Perinatal Discovery, University of California San Diego, La Jolla, CA 92093, USA
| | - Harneet Arora
- 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
- Center for Perinatal Discovery, University of California San Diego, La Jolla, CA 92093, USA
| | - Madeline G. Jackson
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Center for Perinatal Discovery, University of California San Diego, La Jolla, CA 92093, USA
| | - Donald Pizzo
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA
| | - Thorold W. Theunissen
- Department of Developmental Biology and Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, 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
- Center for Perinatal Discovery, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
13
|
Shannon MJ, McNeill GL, Koksal B, Baltayeva J, Wächter J, Castellana B, Peñaherrera MS, Robinson WP, Leung PCK, Beristain AG. Single-cell assessment of primary and stem cell-derived human trophoblast organoids as placenta-modeling platforms. Dev Cell 2024; 59:776-792.e11. [PMID: 38359834 DOI: 10.1016/j.devcel.2024.01.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 11/15/2023] [Accepted: 01/23/2024] [Indexed: 02/17/2024]
Abstract
Human trophoblast stem cells (hTSCs) and related trophoblast organoids are state-of-the-art culture systems that facilitate the study of trophoblast development and human placentation. Using single-cell transcriptomics, we evaluate how organoids derived from freshly isolated first-trimester trophoblasts or from established hTSC cell lines reproduce developmental cell trajectories and transcriptional regulatory processes defined in vivo. Although organoids from primary trophoblasts and hTSCs overall model trophoblast differentiation with accuracy, specific features related to trophoblast composition, trophoblast differentiation, and transcriptional drivers of trophoblast development show levels of misalignment. This is best illustrated by the identification of an expanded progenitor state in stem cell-derived organoids that is nearly absent in vivo and transcriptionally shares both villous cytotrophoblast and extravillous trophoblast characteristics. Together, this work provides a comprehensive resource that identifies strengths and limitations of current trophoblast organoid platforms.
Collapse
Affiliation(s)
- Matthew J Shannon
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| | - Gina L McNeill
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| | - Burak Koksal
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| | - Jennet Baltayeva
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| | - Jasmin Wächter
- Department of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - Barbara Castellana
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| | - Maria S Peñaherrera
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Medical Genetics, The University of British Columbia, Vancouver, BC, Canada
| | - Wendy P Robinson
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Medical Genetics, The University of British Columbia, Vancouver, BC, Canada
| | - Peter C K Leung
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| | - Alexander G Beristain
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
14
|
Lermant A, Rabussier G, Davidson L, Lanz HL, Murdoch CE. Protocol for a placenta-on-a-chip model using trophoblasts differentiated from human induced pluripotent stem cells. STAR Protoc 2024; 5:102879. [PMID: 38358879 PMCID: PMC10876593 DOI: 10.1016/j.xpro.2024.102879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/07/2023] [Accepted: 01/25/2024] [Indexed: 02/17/2024] Open
Abstract
Organ-on-a-chip technology incorporating stem cell techniques represents a promising strategy to improve modeling of human organs. Here, we present a protocol for generating a standardized 3D placenta-on-a-chip model using trophoblast derived from human induced pluripotent stem cells (hiPSCs). We describe steps for seeding hiPSCs into multi-chip OrganoPlate devices and on-chip differentiation into trophoblasts against an extracellular matrix under perfused conditions. We then detail procedures for conducting a functional barrier integrity assay, immunostaining, and collecting protein or RNA for molecular analysis. For complete details on the use and execution of this protocol, please refer to Lermant et al. (2023).1.
Collapse
Affiliation(s)
- Agathe Lermant
- Systems Medicine, School of Medicine, University of Dundee, DD1 9SY Dundee, UK
| | | | - Lindsey Davidson
- Human Pluripotent Stem Cell Facility, School of Life Sciences, University of Dundee, Dundee, UK
| | | | - Colin E Murdoch
- Systems Medicine, School of Medicine, University of Dundee, DD1 9SY Dundee, UK.
| |
Collapse
|
15
|
Anvar Z, Chakchouk I, Sharif M, Mahadevan S, Su L, Anikar S, Naini FA, Utama AB, Van den Veyver IB. Comparison of Four Protocols for In Vitro Differentiation of Human Embryonic Stem Cells into Trophoblast Lineages by BMP4 and Dual Inhibition of Activin/Nodal and FGF2 Signaling. Reprod Sci 2024; 31:173-189. [PMID: 37658178 PMCID: PMC10784360 DOI: 10.1007/s43032-023-01334-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 08/16/2023] [Indexed: 09/03/2023]
Abstract
Human embryonic stem cells (hESCs) cultured in media containing bone morphogenic protein 4 (BMP4; B) differentiate into trophoblast-like cells. Supplementing media with inhibitors of activin/nodal signaling (A83-01) and of fibroblast growth factor 2 (PD173074) suppresses mesoderm and endoderm formation and improves specification of trophoblast-like lineages, but with variable effectiveness. We compared differentiation in four BMP4-containing media: mTeSR1-BMP4 only, mTeSR1-BAP, basal medium with BAP (basal-BAP), and a newly defined medium, E7-BAP. These media variably drive early differentiation towards trophoblast-like lineages with upregulation of early trophoblast markers CDX2 and KRT7 and downregulation of pluripotency markers (OCT4 and NANOG). As expected, based on differences between media in FGF2 and its inhibitors, downregulation of mesendoderm marker EOMES was variable between media. By day 7, only hESCs grown in E7-BAP or basal-BAP expressed HLA-G protein, indicating the presence of cells with extravillous trophoblast characteristics. Expression of HLA-G and other differentiation markers (hCG, KRT7, and GCM1) was highest in basal-BAP, suggesting a faster differentiation in this medium, but those cultures were more inhomogeneous and still expressed some endodermal and pluripotency markers. In E7-BAP, HLA-G expression increased later and was lower. There was also a low but maintained expression of some C19MC miRNAs, with more CpG hypomethylation of the ELF5 promoter, suggesting that E7-BAP cultures differentiate slower along the trophoblast lineage. We conclude that while all protocols drive differentiation into trophoblast lineages with varying efficiency, they have advantages and disadvantages that must be considered when selecting a protocol for specific experiments.
Collapse
Affiliation(s)
- Zahra Anvar
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
- Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Room 1025.14, Houston, TX, 77030, USA
| | - Imen Chakchouk
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
- Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Room 1025.14, Houston, TX, 77030, USA
| | - Momal Sharif
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
- Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Room 1025.14, Houston, TX, 77030, USA
| | - Sangeetha Mahadevan
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
- Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Room 1025.14, Houston, TX, 77030, USA
| | - Li Su
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
- Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Room 1025.14, Houston, TX, 77030, USA
| | - Swathi Anikar
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
- Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Room 1025.14, Houston, TX, 77030, USA
| | - Fatemeh Alavi Naini
- Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Room 1025.14, Houston, TX, 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | | | - Ignatia B Van den Veyver
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA.
- Duncan Neurological Research Institute, Texas Children's Hospital, 1250 Moursund Street, Room 1025.14, Houston, TX, 77030, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
16
|
Dhanjal DS, Singh R, Sharma V, Nepovimova E, Adam V, Kuca K, Chopra C. Advances in Genetic Reprogramming: Prospects from Developmental Biology to Regenerative Medicine. Curr Med Chem 2024; 31:1646-1690. [PMID: 37138422 DOI: 10.2174/0929867330666230503144619] [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: 11/12/2022] [Revised: 03/13/2023] [Accepted: 03/16/2023] [Indexed: 05/05/2023]
Abstract
The foundations of cell reprogramming were laid by Yamanaka and co-workers, who showed that somatic cells can be reprogrammed into pluripotent cells (induced pluripotency). Since this discovery, the field of regenerative medicine has seen advancements. For example, because they can differentiate into multiple cell types, pluripotent stem cells are considered vital components in regenerative medicine aimed at the functional restoration of damaged tissue. Despite years of research, both replacement and restoration of failed organs/ tissues have remained elusive scientific feats. However, with the inception of cell engineering and nuclear reprogramming, useful solutions have been identified to counter the need for compatible and sustainable organs. By combining the science underlying genetic engineering and nuclear reprogramming with regenerative medicine, scientists have engineered cells to make gene and stem cell therapies applicable and effective. These approaches have enabled the targeting of various pathways to reprogramme cells, i.e., make them behave in beneficial ways in a patient-specific manner. Technological advancements have clearly supported the concept and realization of regenerative medicine. Genetic engineering is used for tissue engineering and nuclear reprogramming and has led to advances in regenerative medicine. Targeted therapies and replacement of traumatized , damaged, or aged organs can be realized through genetic engineering. Furthermore, the success of these therapies has been validated through thousands of clinical trials. Scientists are currently evaluating induced tissue-specific stem cells (iTSCs), which may lead to tumour-free applications of pluripotency induction. In this review, we present state-of-the-art genetic engineering that has been used in regenerative medicine. We also focus on ways that genetic engineering and nuclear reprogramming have transformed regenerative medicine and have become unique therapeutic niches.
Collapse
Affiliation(s)
- Daljeet Singh Dhanjal
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Reena Singh
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Varun Sharma
- Head of Bioinformatic Division, NMC Genetics India Pvt. Ltd., Gurugram, India
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 50003, Czech Republic
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, Brno, CZ 613 00, Czech Republic
- Central European Institute of Technology, Brno University of Technology, Purkynova 123, Brno, CZ-612 00, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 50003, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, 50005, Czech Republic
| | - Chirag Chopra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
17
|
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: 1.0] [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
|
18
|
Chehelgerdi M, Behdarvand Dehkordi F, Chehelgerdi M, Kabiri H, Salehian-Dehkordi H, Abdolvand M, Salmanizadeh S, Rashidi M, Niazmand A, Ahmadi S, Feizbakhshan S, Kabiri S, Vatandoost N, Ranjbarnejad T. Exploring the promising potential of induced pluripotent stem cells in cancer research and therapy. Mol Cancer 2023; 22:189. [PMID: 38017433 PMCID: PMC10683363 DOI: 10.1186/s12943-023-01873-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/27/2023] [Indexed: 11/30/2023] Open
Abstract
The advent of iPSCs has brought about a significant transformation in stem cell research, opening up promising avenues for advancing cancer treatment. The formation of cancer is a multifaceted process influenced by genetic, epigenetic, and environmental factors. iPSCs offer a distinctive platform for investigating the origin of cancer, paving the way for novel approaches to cancer treatment, drug testing, and tailored medical interventions. This review article will provide an overview of the science behind iPSCs, the current limitations and challenges in iPSC-based cancer therapy, the ethical and social implications, and the comparative analysis with other stem cell types for cancer treatment. The article will also discuss the applications of iPSCs in tumorigenesis, the future of iPSCs in tumorigenesis research, and highlight successful case studies utilizing iPSCs in tumorigenesis research. The conclusion will summarize the advancements made in iPSC-based tumorigenesis research and the importance of continued investment in iPSC research to unlock the full potential of these cells.
Collapse
Affiliation(s)
- Matin Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Fereshteh Behdarvand Dehkordi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Mohammad Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran.
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Hamidreza Kabiri
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | | | - Mohammad Abdolvand
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Sharareh Salmanizadeh
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Hezar-Jereeb Street, Isfahan, 81746-73441, Iran
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Anoosha Niazmand
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Saba Ahmadi
- Department of Molecular and Medical Genetics, Tbilisi State Medical University, Tbilisi, Georgia
| | - Sara Feizbakhshan
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Saber Kabiri
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Nasimeh Vatandoost
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Tayebeh Ranjbarnejad
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| |
Collapse
|
19
|
Liu X, Wang G, Huang H, Lv X, Si Y, Bai L, Wang G, Li Q, Yang W. Exploring maternal-fetal interface with in vitro placental and trophoblastic models. Front Cell Dev Biol 2023; 11:1279227. [PMID: 38033854 PMCID: PMC10682727 DOI: 10.3389/fcell.2023.1279227] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
The placenta, being a temporary organ, plays a crucial role in facilitating the exchange of nutrients and gases between the mother and the fetus during pregnancy. Any abnormalities in the development of this vital organ not only lead to various pregnancy-related disorders that can result in fetal injury or death, but also have long-term effects on maternal health. In vitro models have been employed to study the physiological features and molecular regulatory mechanisms of placental development, aiming to gain a detailed understanding of the pathogenesis of pregnancy-related diseases. Among these models, trophoblast stem cell culture and organoids show great promise. In this review, we provide a comprehensive overview of the current mature trophoblast stem cell models and emerging organoid models, while also discussing other models in a systematic manner. We believe that this knowledge will be valuable in guiding further exploration of the complex maternal-fetal interface.
Collapse
Affiliation(s)
- Xinlu Liu
- School of Biosciences and Biotechnology, Weifang Medical University, Weifang, Shandong, China
| | - Gang Wang
- Department of Emergency, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Haiqin Huang
- School of Biosciences and Biotechnology, Weifang Medical University, Weifang, Shandong, China
| | - Xin Lv
- School of Biosciences and Biotechnology, Weifang Medical University, Weifang, Shandong, China
| | - Yanru Si
- School of Biosciences and Biotechnology, Weifang Medical University, Weifang, Shandong, China
| | - Lixia Bai
- School of Biosciences and Biotechnology, Weifang Medical University, Weifang, Shandong, China
| | - Guohui Wang
- School of Biosciences and Biotechnology, Weifang Medical University, Weifang, Shandong, China
| | - Qinghua Li
- School of Public Health, Weifang Medical University, Weifang, Shandong, China
| | - Weiwei Yang
- School of Biosciences and Biotechnology, Weifang Medical University, Weifang, Shandong, China
| |
Collapse
|
20
|
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: 19] [Impact Index Per Article: 9.5] [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
|
21
|
Cheung VC, Bui T, Soncin F, Bai T, Kessler JA, Parast MM, Horii M. Current Strategies of Modeling Human Trophoblast Using Human Pluripotent Stem Cells in vitro. Curr Protoc 2023; 3:e875. [PMID: 37787612 PMCID: PMC10558083 DOI: 10.1002/cpz1.875] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
We previously established a trophoblast differentiation protocol from primed human pluripotent stem cells (PSC). To induce this lineage, we use a combination of Bone Morphogenetic Protein-4 (BMP4) and the WNT inhibitor IWP2. This protocol has enabled us to obtain a pure population of trophectoderm (TE)-like cells that could subsequently be terminally differentiated into syncytiotrophoblasts (STB) and extravillous trophoblasts (EVT). However, the resulting TE-like cells could only be terminally differentiated to a variable mixture of STB and EVT, with a bias toward the STB lineage. Recently, methods have been developed for derivation and culture of self-renewing human trophoblast stem cells (TSC) from human embryos and early gestation placental tissues. These primary TSCs were further able to differentiate into either STB or EVT with high efficiency using the lineage specific differentiation protocols. Based partly on these protocols, we have developed methods for establishing self-renewing TSC-like cells from PSC, and for efficient lineage-specific terminal differentiation. Here, we describe in detail the protocols to derive and maintain PSC-TSC, from both embryonic stem cells (ESC) and patient-derived induced pluripotent stem cells (iPSC), and their subsequent terminal differentiation to STB and EVT. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Trophoblast Differentiation into TE-like Cells Basic Protocol 2: Conversion of PSC-Derived TE-like Cells to TSC Basic Protocol 3: Passaging PSC-Derived TSC in iCTB Complete Medium Basic Protocol 4: STB Differentiation from PSC-derived TSC Basic Protocol 5: EVT Differentiation from PSC-derived TSC Support Protocol 1: Geltrex-coated tissue culture plate preparation Support Protocol 2: Collagen IV-coated tissue culture plate preparation Support Protocol 3: Fibronectin-coated tissue culture plate preparation.
Collapse
Affiliation(s)
- Virginia Chu Cheung
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - 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
| | - Francesca Soncin
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Tao Bai
- Department of Neurology, Northwestern University, Chicago, IL 60611, USA
| | - John A. Kessler
- Department of Neurology, Northwestern University, Chicago, IL 60611, USA
| | - Mana M Parast
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - 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
|
22
|
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: 2.0] [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
|
23
|
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: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [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
|
24
|
Ai Z, Niu B, Yin Y, Xiang L, Shi G, Duan K, Wang S, Hu Y, Zhang C, Zhang C, Rong L, Kong R, Chen T, Guo Y, Liu W, Li N, Zhao S, Zhu X, Mai X, Li Y, Wu Z, Zheng Y, Fu J, Ji W, Li T. Dissecting peri-implantation development using cultured human embryos and embryo-like assembloids. Cell Res 2023; 33:661-678. [PMID: 37460804 PMCID: PMC10474050 DOI: 10.1038/s41422-023-00846-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 06/24/2023] [Indexed: 09/03/2023] Open
Abstract
Studies of cultured embryos have provided insights into human peri-implantation development. However, detailed knowledge of peri-implantation lineage development as well as underlying mechanisms remains obscure. Using 3D-cultured human embryos, herein we report a complete cell atlas of the early post-implantation lineages and decipher cellular composition and gene signatures of the epiblast and hypoblast derivatives. In addition, we develop an embryo-like assembloid (E-assembloid) by assembling naive hESCs and extraembryonic cells. Using human embryos and E-assembloids, we reveal that WNT, BMP and Nodal signaling pathways synergistically, but functionally differently, orchestrate human peri-implantation lineage development. Specially, we dissect mechanisms underlying extraembryonic mesoderm and extraembryonic endoderm specifications. Finally, an improved E-assembloid is developed to recapitulate the epiblast and hypoblast development and tissue architectures in the pre-gastrulation human embryo. Our findings provide insights into human peri-implantation development, and the E-assembloid offers a useful model to disentangle cellular behaviors and signaling interactions that drive human embryogenesis.
Collapse
Affiliation(s)
- Zongyong Ai
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China.
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China.
- Yunnan Provincial Academy of Science and Technology, Kunming, Yunnan, China.
| | - Baohua Niu
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Yu Yin
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
- Yunnan Provincial Academy of Science and Technology, Kunming, Yunnan, China
| | - Lifeng Xiang
- Department of Reproductive Medicine, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Gaohui Shi
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Kui Duan
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
- Yunnan Provincial Academy of Science and Technology, Kunming, Yunnan, China
| | - Sile Wang
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
- Yunnan Provincial Academy of Science and Technology, Kunming, Yunnan, China
| | - Yingjie Hu
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Chi Zhang
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Chengting Zhang
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
- Yunnan Provincial Academy of Science and Technology, Kunming, Yunnan, China
| | - Lujuan Rong
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Ruize Kong
- Department of Reproductive Medicine, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Tingwei Chen
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Yixin Guo
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, International Campus, Zhejiang University, Haining, Zhejiang, China
| | - Wanlu Liu
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, International Campus, Zhejiang University, Haining, Zhejiang, China
| | - Nan Li
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Shumei Zhao
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
- Yunnan Provincial Academy of Science and Technology, Kunming, Yunnan, China
| | - Xiaoqing Zhu
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
- Yunnan Provincial Academy of Science and Technology, Kunming, Yunnan, China
| | - Xuancheng Mai
- Department of Reproductive Medicine, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Yonggang Li
- Department of Reproductive Medicine, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Ze Wu
- Department of Reproductive Medicine, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Yi Zheng
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Weizhi Ji
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China.
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China.
- Yunnan Provincial Academy of Science and Technology, Kunming, Yunnan, China.
| | - Tianqing Li
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China.
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China.
- Yunnan Provincial Academy of Science and Technology, Kunming, Yunnan, China.
| |
Collapse
|
25
|
Lermant A, Rabussier G, Lanz HL, Davidson L, Porter IM, Murdoch CE. Development of a human iPSC-derived placental barrier-on-chip model. iScience 2023; 26:107240. [PMID: 37534160 PMCID: PMC10392097 DOI: 10.1016/j.isci.2023.107240] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/28/2023] [Accepted: 06/26/2023] [Indexed: 08/04/2023] Open
Abstract
Although recently developed placenta-on-chip systems opened promising perspectives for placental barrier modeling, they still lack physiologically relevant trophoblasts and are poorly amenable to high-throughput studies. We aimed to implement human-induced pluripotent stem cells (hiPSC)-derived trophoblasts into a multi-well microfluidic device to develop a physiologically relevant and scalable placental barrier model. When cultured in a perfused micro-channel against a collagen-based matrix, hiPSC-derived trophoblasts self-arranged into a 3D structure showing invasive behavior, fusogenic and endocrine activities, structural integrity, and expressing placental transporters. RNA-seq analysis revealed that the microfluidic 3D environment boosted expression of genes related to early placental structural development, mainly involved in mechanosensing and cell surface receptor signaling. These results demonstrated the feasibility of generating a differentiated primitive syncytium from hiPSC in a microfluidic platform. Besides expanding hiPSC-derived trophoblast scope of applications, this study constitutes an important resource to improve placental barrier models and boost research and therapeutics evaluation in pregnancy.
Collapse
Affiliation(s)
- Agathe Lermant
- Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| | | | | | - Lindsay Davidson
- Human Pluripotent Stem Cell Facility, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Iain M. Porter
- Dundee Imaging Facility, School of Life Sciences, University of Dundee, DD1 5EH, UK
| | - Colin E. Murdoch
- Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| |
Collapse
|
26
|
Jaremek A, Shaha S, Jeyarajah MJ, Jaju Bhattad G, Chowdhury D, Riddell M, Renaud SJ. Genome-Wide Analysis of Hypoxia-Inducible Factor Binding Reveals Targets Implicated in Impaired Human Placental Syncytiotrophoblast Formation under Low Oxygen. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:846-865. [PMID: 37028593 DOI: 10.1016/j.ajpath.2023.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 01/31/2023] [Accepted: 03/16/2023] [Indexed: 04/08/2023]
Abstract
Preeclampsia (PE) is a common and serious complication of pregnancy with no cure except premature delivery. The root cause of PE is improper development of the placenta-the temporary organ supporting fetal growth and development. Continuous formation of the multinucleated syncytiotrophoblast (STB) layer via differentiation and fusion of cytotrophoblasts (CTBs) is vital for healthy placentation and is impaired in preeclamptic pregnancies. In PE, there is reduced/intermittent placental perfusion, likely resulting in a persistently low O2 environment. Low O2 inhibits differentiation and fusion of CTBs into STB and may thus contribute to PE pathogenesis; however, the underlying mechanisms are unknown. Because low O2 activates a transcription factor complex in cells known as the hypoxia-inducible factor (HIF), the objective of this study was to investigate whether HIF signaling inhibits STB formation by regulating genes required for this process. Culture of primary CTBs, the CTB-like cell line BeWo, and human trophoblast stem cells under low O2 reduced cell fusion and differentiation into STB. Knockdown of aryl hydrocarbon receptor nuclear translocator (a key component of the HIF complex) in BeWo cells restored syncytialization and expression of STB-associated genes under different O2 levels. Chromatin immunoprecipitation sequencing facilitated the identification of global aryl hydrocarbon receptor nuclear translocator/HIF binding sites, including several near genes implicated in STB development, such as ERVH48-1 and BHLHE40, providing new insights into mechanisms underlying pregnancy diseases linked to poor placental O2 supply.
Collapse
Affiliation(s)
- Adam Jaremek
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Sumaiyah Shaha
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | - Mariyan J Jeyarajah
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Gargi Jaju Bhattad
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Diba Chowdhury
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Meghan Riddell
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada; Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
| | - Stephen J Renaud
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; Children's Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada.
| |
Collapse
|
27
|
Pennarossa G, Arcuri S, De Iorio T, Ledda S, Gandolfi F, Brevini TAL. Combination of epigenetic erasing and mechanical cues to generate human epiBlastoids from adult dermal fibroblasts. J Assist Reprod Genet 2023; 40:1015-1027. [PMID: 36933093 PMCID: PMC10024007 DOI: 10.1007/s10815-023-02773-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 03/09/2023] [Indexed: 03/19/2023] Open
Abstract
PURPOSE This study is to develop a new protocol that combines the use of epigenetic cues and mechanical stimuli to assemble 3D spherical structures, arbitrarily defined "epiBlastoids," whose phenotype is remarkably similar to natural embryos. METHODS A 3-step approach is used to generate epiBlastoids. In the first step, adult dermal fibroblasts are converted into trophoblast (TR)-like cells, combining the use of 5-azacytidine, to erase the original phenotype, with an ad hoc induction protocol, to drive cells towards TR lineage. In the second step, epigenetic erasing is applied once again, in combination with mechanosensing-related cues, to generate inner cell mass (ICM)-like organoids. Specifically, erased cells are encapsulated into micro-bioreactors to promote 3D cell rearrangement and boost pluripotency. In the third step, TR-like cells are co-cultured with ICM-like spheroids in the same micro-bioreactors. Subsequently, the newly generated embryoids are transferred to microwells to favor epiBlastoid formation. RESULTS Adult dermal fibroblasts are successfully readdressed towards TR lineage. Cells subjected to epigenetic erasing and encapsulated into micro-bioreactors rearrange in 3D ICM-like structures. Co-culture of TR-like cells and ICM-like spheroids into micro-bioreactors and microwells induces the formation of single structures with uniform shape reminiscent in vivo embryos. CDX2+ cells localized in the out layer of the spheroids, while OCT4+ cells in the inner of the structures. TROP2+ cells display YAP nuclear accumulation and actively transcribed for mature TR markers, while TROP2- cells showed YAP cytoplasmic compartmentalization and expressed pluripotency-related genes. CONCLUSION We describe the generation of epiBlastoids that may find useful application in the assisted reproduction field.
Collapse
Affiliation(s)
- Georgia Pennarossa
- Department of Veterinary Medicine and Animal Science, Center for Stem Cell Research, Laboratory of Biomedical Embryology and Tissue Engineering, Università Degli Studi Di Milano, 26900, Lodi, Italy
| | - Sharon Arcuri
- Department of Veterinary Medicine and Animal Science, Center for Stem Cell Research, Laboratory of Biomedical Embryology and Tissue Engineering, Università Degli Studi Di Milano, 26900, Lodi, Italy
| | - Teresina De Iorio
- Department of Veterinary Medicine and Animal Science, Center for Stem Cell Research, Laboratory of Biomedical Embryology and Tissue Engineering, Università Degli Studi Di Milano, 26900, Lodi, Italy
| | - Sergio Ledda
- Department of Veterinary Medicine, University of Sassari, 07100, Sassari, Italy
| | - Fulvio Gandolfi
- Department of Agricultural and Environmental Sciences - Production, Landscape, Agroenergy, Università degli Studi di Milano, 20133, Milan, Italy
| | - Tiziana A L Brevini
- Department of Veterinary Medicine and Animal Science, Center for Stem Cell Research, Laboratory of Biomedical Embryology and Tissue Engineering, Università Degli Studi Di Milano, 26900, Lodi, Italy.
| |
Collapse
|
28
|
Rabussier G, Bünter I, Bouwhuis J, Soragni C, van Zijp T, Ng CP, Domansky K, de Windt LJ, Vulto P, Murdoch CE, Bircsak KM, Lanz HL. Healthy and diseased placental barrier on-a-chip models suitable for standardized studies. Acta Biomater 2023; 164:363-376. [PMID: 37116636 DOI: 10.1016/j.actbio.2023.04.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 04/05/2023] [Accepted: 04/21/2023] [Indexed: 04/30/2023]
Abstract
Pathologies associated with uteroplacental hypoxia, such as preeclampsia are among the leading causes of maternal and perinatal morbidity in the world. Its fundamental mechanisms are yet poorly understood due to a lack of good experimental models. Here we report an in vitro model of the placental barrier, based on co-culture of trophoblasts and endothelial cells against a collagen extracellular matrix in a microfluidic platform. The model yields a functional syncytium with barrier properties, polarization, secretion of relevant extracellular membrane components, thinning of the materno-fetal space, hormone secretion, and transporter function. The model is exposed to low oxygen conditions and perfusion flow is modulated to induce a pathological environment. This results in reduced barrier function, hormone secretion, and microvilli as well as an increased nuclei count, characteristics of preeclamptic placentas. The model is implemented in a titer plate-based microfluidic platform fully amenable to high-throughput screening. We thus believe this model could aid mechanistic understanding of preeclampsia and other placental pathologies associated with hypoxia/ischemia, as well as support future development of effective therapies through target and compound screening campaigns. STATEMENT OF SIGNIFICANCE: : The human placenta is a unique organ sustaining fetus growth but is also the source of severe pathologies, such as Preeclampsia. Though leading cause of perinatal mortality in the world, preeclampsia remains untreatable due to a lack of relevant in vitro placenta models. To better understand the pathology, we have developed 3D placental barrier models in a microfluidic device. The platform allows parallel culture of 40 perfused physiological miniaturized placental barriers, comprising a differentiated syncytium and endothelium that have been validated for transporter functions. Exposure to a hypoxic and ischemic environment enabled the mimicking of preeclamptic characteristics in high-throughput, which we believe could lead to a better understanding of the pathology as well as support future effective therapies development.
Collapse
Affiliation(s)
- Gwenaëlle Rabussier
- MIMETAS BV, Oegstgeest, 2342 DH, The Netherlands; Department of Cardiology, Maastricht University, Maastricht, 6226 ER, The Netherlands
| | - Ivan Bünter
- MIMETAS BV, Oegstgeest, 2342 DH, The Netherlands
| | | | - Camilla Soragni
- MIMETAS BV, Oegstgeest, 2342 DH, The Netherlands; Department of Cardiology, Maastricht University, Maastricht, 6226 ER, The Netherlands
| | | | - Chee Ping Ng
- MIMETAS BV, Oegstgeest, 2342 DH, The Netherlands
| | | | - Leon J de Windt
- Department of Cardiology, Maastricht University, Maastricht, 6226 ER, The Netherlands
| | - Paul Vulto
- MIMETAS BV, Oegstgeest, 2342 DH, The Netherlands
| | - Colin E Murdoch
- Systems Medicine, School of Medicine, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | | | | |
Collapse
|
29
|
Sonnemann HM, Pazdrak B, Antunes DA, Roszik J, Lizée G. Vestigial-like 1 (VGLL1): An ancient co-transcriptional activator linking wing, placenta, and tumor development. Biochim Biophys Acta Rev Cancer 2023; 1878:188892. [PMID: 37004960 DOI: 10.1016/j.bbcan.2023.188892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
Vestigial-like 1 (VGLL1) is a recently discovered driver of proliferation and invasion that is expressed in many aggressive human malignancies and is strongly associated with poor prognosis. The VGLL1 gene encodes for a co-transcriptional activator that shows intriguing structural similarity to key activators in the hippo pathway, providing important clues to its functional role. VGLL1 binds to TEADs in an analogous fashion to YAP1 but appears to activate a distinct set of downstream gene targets. In mammals, VGLL1 expression is found almost exclusively in placental trophoblasts, cells that share many hallmarks of cancer. Due to its role as a driver of tumor progression, VGLL1 has become a target of interest for potential anticancer therapies. In this review, we discuss VGLL1 from an evolutionary perspective, contrast its role in placental and tumor development, summarize the current knowledge of how signaling pathways can modulate VGLL1 function, and discuss potential approaches for targeting VGLL1 therapeutically.
Collapse
|
30
|
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: 3.5] [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
|
31
|
Castel G, David L. Induction of human trophoblast stem cells. Nat Protoc 2022; 17:2760-2783. [PMID: 36241723 DOI: 10.1038/s41596-022-00744-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 06/22/2022] [Indexed: 02/07/2023]
Abstract
Cell reprogramming has allowed unprecedented access to human development, from virtually any genome. However, reprogramming yields pluripotent stem cells that can differentiate into all cells that form a fetus, but not extraembryonic annexes. Therefore, a cellular model allowing study of placental development from a broad genomic repertoire is lacking. Here, we describe an optimized protocol to reprogram somatic cells into human induced trophoblast stem cells (hiTSCs) and convert pluripotent stem cells into human converted TSCs (hcTSCs). This protocol enables much-needed genome-specific placental disease modeling. We also detail extravillous trophoblast and syncytiotrophoblast differentiation protocols from hiTSCs and hcTSCs, a necessary step to validate these cells. In total, this protocol takes 4 months and requires advanced cell culture skills, comparable to those necessary for somatic cell reprogramming into human induced pluripotent stem cells.
Collapse
Affiliation(s)
- Gaël Castel
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Laurent David
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France.
- Nantes Université, CHU Nantes, INSERM, CNRS, BioCore, Nantes, France.
| |
Collapse
|
32
|
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: 16] [Impact Index Per Article: 5.3] [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
|
33
|
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: 2.0] [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
|
34
|
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: 16] [Impact Index Per Article: 5.3] [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
|
35
|
Zhang Y, An C, Yu Y, Lin J, Jin L, Li C, Tan T, Yu Y, Fan Y. Epidermal growth factor induces a trophectoderm lineage transcriptome resembling that of human embryos during reconstruction of blastoids from extended pluripotent stem cells. Cell Prolif 2022; 55:e13317. [PMID: 35880490 PMCID: PMC9628219 DOI: 10.1111/cpr.13317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/23/2022] [Accepted: 06/27/2022] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVES This study aims to optimize the human extended pluripotent stem cell (EPSC) to trophectoderm (TE)-like cell induction with addition of EGF and improve the quality of the reconstructing blastoids. MATERIALS AND METHODS TE-like cells were differentiated from human EPSCs. RNA-seq data analysis was performed to compare with TE-like cells from multiple human pluripotent stem cells (hPSCs) and embryos. A small-scale compound selection was performed for optimizing the TE-like cell induction and the efficiency was characterized using TE-lineage markers expression by immunofluorescence stanning. Blastoids were generated by using the optimized TE-like cells and the undifferentiated human EPSCs through three-dimensional culture system. Single-cell RNA sequencing was performed to investigate the lineage segregation of the optimized blastoids to human blastocysts. RESULTS TE-like cells derived from human EPSCs exhibited similar transcriptome with TE cells from embryos. Additionally, TE-like cells from multiple naive hPSCs exhibited heterogeneous gene expression patterns and signalling pathways because of the incomplete silencing of naive-specific genes and loss of imprinting. Furthermore, with the addition of EGF, TE-like cells derived from human EPSCs enhanced the TE lineage-related signalling pathways and exhibited more similar transcriptome to human embryos. Through resembling with undifferentiated human EPSCs, we elevated the quality and efficiency of reconstructing blastoids and separated more lineage cells with precise temporal and spatial expression, especially the PE lineage. CONCLUSION Addition of EGF enhanced TE lineage differentiation and human blastoids reconstruction. The optimized blastoids could be used as a blastocyst model for simulating early embryonic development.
Collapse
Affiliation(s)
- Yingying Zhang
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education InstitutesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Chenrui An
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education InstitutesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Yanhong Yu
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education InstitutesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Jiajing Lin
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education InstitutesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Long Jin
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education InstitutesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Chaohui Li
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education InstitutesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Tao Tan
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
| | - Yang Yu
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center for Reproductive Medicine, Department of Obstetrics and GynecologyPeking University Third HospitalBeijingChina
| | - Yong Fan
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education InstitutesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| |
Collapse
|
36
|
Monosomy X in isogenic human iPSC-derived trophoblast model impacts expression modules preserved in human placenta. Proc Natl Acad Sci U S A 2022; 119:e2211073119. [PMID: 36161909 DOI: 10.1073/pnas.2211073119] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mammalian sex chromosomes encode homologous X/Y gene pairs that were retained on the Y chromosome in males and escape X chromosome inactivation (XCI) in females. Inferred to reflect X/Y pair dosage sensitivity, monosomy X is a leading cause of miscarriage in humans with near full penetrance. This phenotype is shared with many other mammals but not the mouse, which offers sophisticated genetic tools to generate sex chromosomal aneuploidy but also tolerates its developmental impact. To address this critical gap, we generated X-monosomic human induced pluripotent stem cells (hiPSCs) alongside otherwise isogenic euploid controls from male and female mosaic samples. Phased genomic variants in these hiPSC panels enable systematic investigation of X/Y dosage-sensitive features using in vitro models of human development. Here, we demonstrate the utility of these validated hiPSC lines to test how X/Y-linked gene dosage impacts a widely used model for human syncytiotrophoblast development. While these isogenic panels trigger a GATA2/3- and TFAP2A/C-driven trophoblast gene circuit irrespective of karyotype, differential expression implicates monosomy X in altered levels of placental genes and in secretion of placental growth factor (PlGF) and human chorionic gonadotropin (hCG). Remarkably, weighted gene coexpression network modules that significantly reflect these changes are also preserved in first-trimester chorionic villi and term placenta. Our results suggest monosomy X may skew trophoblast cell type composition and function, and that the combined haploinsufficiency of the pseudoautosomal region likely plays a key role in these changes.
Collapse
|
37
|
Serudji J, Irawati N, Mose JC, Ali H, Yusrawati Y. Serum HIF-1α Levels, miR-210 Expressions, and Reactive Oxygen Species Levels in Early Abortion and Normal Pregnancy. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.9325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background: The blastocyst implants in a relatively hypoxic state. Hypoxic state triggers hypoxia-inducible factor-1α (HIF-1ɑ) production, upregulates the transcription factor miR-210, and stimulates reactive oxygen species (ROS) production by trophoblast cells. HIF-1α also increases the expression of miR-210. High expression of micro-RNA 210 (miR-210) suppresses mitochondrial respiration, increasing ROS production. High level of ROS may result in DNA damage or cell disfunction, thereby impaired trophoblast invasion, leading to early abortion. This study aims to determine the differences of serum HIF-1ɑ levels, miR-210 expressions, and ROS levels between early abortion and normal pregnancy.
Method: This cross-sectional comparative study was conducted in Dr. M. Djamil Hospital Padang, Andalas University Hospital, and 5 Public Health Centers in Padang. Fifty-patients with gestational age less than 12 weeks (25 early abortions and 25 normal pregnancies) were included in this study. All samples were tested for HIF-1ɑ and ROS level using enzyme-linked immunosorbent assay (ELISA) method, and miR-210 expression using real-time polymerase chain reaction (PCR) technique. Spearman correlation and Mann Whitney test. was used in this study.
Results: Both study groups were equivalent in terms of age, gestational age, and gravidity (p = 0.51, 0.453 and 1.00). The median of HIF-1ɑ level, miR-210 expression, and ROS level were higher in early abortions than normal pregnancies i.e (3.73 vs 3.42) ng/mL (p = 0.016), (43.55 vs 17.85) copies/ng (p = 0.027), and (1.36 vs 1.20) ng/mL (p = 0.003). The coefficient correlations were 0.16 between HIF-1ɑ level and miR-210 expression (p=0.267), 0.46 between HIF-1ɑ level and ROS level (p=0.001), and 0.18 between miR-210 expression and ROS level (p=0.207).
Conclusion: HIF-1ɑ level, miR-210 expression, and ROS level were associated with early abortion. HIF-1ɑ level has a correlation with ROS level.
Collapse
|
38
|
Roberts RM, Ezashi T, Temple J, Owen JR, Soncin F, Parast MM. The role of BMP4 signaling in trophoblast emergence from pluripotency. Cell Mol Life Sci 2022; 79:447. [PMID: 35877048 PMCID: PMC10243463 DOI: 10.1007/s00018-022-04478-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/24/2022] [Accepted: 07/06/2022] [Indexed: 11/03/2022]
Abstract
The Bone Morphogenetic Protein (BMP) signaling pathway has established roles in early embryonic morphogenesis, particularly in the epiblast. More recently, however, it has also been implicated in development of extraembryonic lineages, including trophectoderm (TE), in both mouse and human. In this review, we will provide an overview of this signaling pathway, with a focus on BMP4, and its role in emergence and development of TE in both early mouse and human embryogenesis. Subsequently, we will build on these in vivo data and discuss the utility of BMP4-based protocols for in vitro conversion of primed vs. naïve pluripotent stem cells (PSC) into trophoblast, and specifically into trophoblast stem cells (TSC). PSC-derived TSC could provide an abundant, reproducible, and ethically acceptable source of cells for modeling placental development.
Collapse
Affiliation(s)
- R Michael Roberts
- Division of Animal Sciences and Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Toshihiko Ezashi
- Division of Animal Sciences and Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
- Colorado Center for Reproductive Medicine, 10290 Ridgegate Circle, Lone Tree, CO, 80124, USA
| | - Jasmine Temple
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA
| | - Joseph R Owen
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Francesca Soncin
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA
| | - Mana M Parast
- Department of Pathology, University of California San Diego, La Jolla, CA, USA.
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA, 92037, USA.
| |
Collapse
|
39
|
Kojima J, Ono M, Kuji N, Nishi H. Human Chorionic Villous Differentiation and Placental Development. Int J Mol Sci 2022; 23:8003. [PMID: 35887349 PMCID: PMC9325306 DOI: 10.3390/ijms23148003] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/19/2022] [Accepted: 07/19/2022] [Indexed: 02/04/2023] Open
Abstract
In humans, the placenta provides the only fetomaternal connection and is essential for establishing a pregnancy as well as fetal well-being. Additionally, it allows maternal physiological adaptation and embryonic immunological acceptance, support, and nutrition. The placenta is derived from extra-embryonic tissues that develop rapidly and dynamically in the first weeks of pregnancy. It is primarily composed of trophoblasts that differentiate into villi, stromal cells, macrophages, and fetal endothelial cells (FEC). Placental differentiation may be closely related to perinatal diseases, including fetal growth retardation (FGR) and hypertensive disorders of pregnancy (HDP), and miscarriage. There are limited findings regarding human chorionic villous differentiation and placental development because conducting in vivo studies is extremely difficult. Placental tissue varies widely among species. Thus, experimental animal findings are difficult to apply to humans. Early villous differentiation is difficult to study due to the small tissue size; however, a detailed analysis can potentially elucidate perinatal disease causes or help develop novel therapies. Artificial induction of early villous differentiation using human embryonic stem (ES) cells/induced pluripotent stem (iPS) cells was attempted, producing normally differentiated villi that can be used for interventional/invasive research. Here, we summarized and correlated early villous differentiation findings and discussed clinical diseases.
Collapse
Affiliation(s)
| | - Masanori Ono
- Department of Obstetrics and Gynecology, Tokyo Medical University, Tokyo 160-0023, Japan; (J.K.); (N.K.); (H.N.)
| | | | | |
Collapse
|
40
|
Deng P, Cui K, Shi Y, Zhu Y, Wang Y, Shao X, Qin J. Fluidic Flow Enhances the Differentiation of Placental Trophoblast-Like 3D Tissue from hiPSCs in a Perfused Macrofluidic Device. Front Bioeng Biotechnol 2022; 10:907104. [PMID: 35845423 PMCID: PMC9280037 DOI: 10.3389/fbioe.2022.907104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/17/2022] [Indexed: 11/21/2022] Open
Abstract
The human placenta serves as a multifunctional organ to maintain the proper development of a fetus. However, our knowledge of the human placenta is limited due to the lack of appropriate experimental models. In this work, we created an in vitro placental trophoblast-like model via self-organization of human induced pluripotent stem cells (hiPSCs) in a perfused 3D culture macrofluidic device. This device allowed cell seeding, in situ trophoblast lineage differentiation, and formation of trophoblast-like tissues from hiPSCs in a biomimetic microenvironment. It incorporated extracellular matrix (ECM) and fluid flow in a single device. After trophoblast lineage differentiation, we were able to generate the 3D clusters with major cell types of the human placenta, including trophoblast progenitor cytotrophoblasts (CTBs), differentiated subtypes, syncytiotrophoblasts (STBs), and extravillous trophoblasts (EVTs) under long-term 3D culture (∼23 days). Moreover, the formed tissues exhibited enhanced expressions of CTB-, STB-, and EVT-related markers at the level of genes and proteins under a dynamic culture compared with static conditions. RNA-seq analysis revealed the higher expression of trophoblast-specific genes in 3D tissues, indicating the essential role of fluid flow to promote the trophoblast differentiation of hiPSCs. The established placental 3D model combined a bioengineering strategy with developmental principles, providing a promising platform for the study of placental biology in a biomimetic microenvironment in health and disease.
Collapse
Affiliation(s)
- Pengwei Deng
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- Division of Biotechnology, Dalian Institute of Chemical Physics, University of Chinese Academy of Sciences, Beijing, China
| | - Kangli Cui
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- Division of Biotechnology, Dalian Institute of Chemical Physics, University of Chinese Academy of Sciences, Beijing, China
| | - Yang Shi
- Dalian Key Laboratory of Reproduction and Mother-child Genetics, Dalian Women and Children’s Medical Group, Dalian, China
| | - Yujuan Zhu
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- Division of Biotechnology, Dalian Institute of Chemical Physics, University of Chinese Academy of Sciences, Beijing, China
| | - Yaqing Wang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Xiaoguang Shao
- Dalian Key Laboratory of Reproduction and Mother-child Genetics, Dalian Women and Children’s Medical Group, Dalian, China
| | - Jianhua Qin
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- Division of Biotechnology, Dalian Institute of Chemical Physics, University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- *Correspondence: Jianhua Qin,
| |
Collapse
|
41
|
Single-cell transcriptional profiling reveals cellular and molecular divergence in human maternal-fetal interface. Sci Rep 2022; 12:10892. [PMID: 35764880 PMCID: PMC9240006 DOI: 10.1038/s41598-022-14516-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/08/2022] [Indexed: 12/19/2022] Open
Abstract
Placenta plays essential role in successful pregnancy, as the most important organ connecting and interplaying between mother and fetus. However, the cellular characteristics and molecular interaction of cell populations within the fetomaternal interface is still poorly understood. Here, we surveyed the single-cell transcriptomic landscape of human full-term placenta and revealed the heterogeneity of cytotrophoblast cell (CTB) and stromal cell (STR) with the fetal/maternal origin consecutively localized from fetal section (FS), middle section (Mid_S) to maternal section (Mat_S) of maternal–fetal interface. Then, we highlighted a subpopulation of CTB, named trophoblast progenitor-like cells (TPLCs) existed in the full-term placenta and mainly distributed in Mid_S, with high expression of a pool of putative cell surface markers. Further, we revealed the putative key transcription factor PRDM6 that might promote the differentiation of endovascular extravillous trophoblast cells (enEVT) by inhibiting cell proliferation, and down-regulation of PRDM6 might lead to an abnormal enEVT differentiation process in PE. Together, our study offers important resources for better understanding of human placenta and stem cell-based therapy, and provides new insights on the study of tissue heterogeneity, the clinical prevention and control of PE as well as the maternal–fetal interface.
Collapse
|
42
|
Nikitina TV, Lebedev IN. Stem Cell-Based Trophoblast Models to Unravel the Genetic Causes of Human Miscarriages. Cells 2022; 11:1923. [PMID: 35741051 PMCID: PMC9221414 DOI: 10.3390/cells11121923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/10/2022] [Accepted: 06/12/2022] [Indexed: 02/01/2023] Open
Abstract
Miscarriage affects approximately 15% of clinically recognized pregnancies, and 1-3% of couples experience pregnancy loss recurrently. Approximately 50-60% of miscarriages result from chromosomal abnormalities, whereas up to 60% of euploid recurrent abortions harbor variants in candidate genes. The growing number of detected genetic variants requires an investigation into their role in adverse pregnancy outcomes. Since placental defects are the main cause of first-trimester miscarriages, the purpose of this review is to provide a survey of state-of-the-art human in vitro trophoblast models that can be used for the functional assessment of specific abnormalities/variants implicated in pregnancy loss. Since 2018, when primary human trophoblast stem cells were first derived, there has been rapid growth in models of trophoblast lineage. It has been found that a proper balance between self-renewal and differentiation in trophoblast progenitors is crucial for the maintenance of pregnancy. Different responses to aneuploidy have been shown in human embryonic and extra-embryonic lineages. Stem cell-based models provide a powerful tool to explore the effect of a specific aneuploidy/variant on the fetus through placental development, which is important, from a clinical point of view, for deciding on the suitability of embryos for transfer after preimplantation genetic testing for aneuploidy.
Collapse
Affiliation(s)
- Tatiana V. Nikitina
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, 634050 Tomsk, Russia;
| | | |
Collapse
|
43
|
Soncin F, Morey R, Bui T, Requena DF, Cheung VC, Kallol S, Kittle R, Jackson MG, Farah O, Chousal J, Meads M, Pizzo D, Horii M, Fisch KM, Parast MM. Derivation of functional trophoblast stem cells from primed human pluripotent stem cells. Stem Cell Reports 2022; 17:1303-1317. [PMID: 35594858 PMCID: PMC9214048 DOI: 10.1016/j.stemcr.2022.04.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 12/25/2022] Open
Abstract
Trophoblast stem cells (TSCs) have recently been derived from human embryos and early-first-trimester placenta; however, aside from ethical challenges, the unknown disease potential of these cells limits their scientific utility. We have previously established a bone morphogetic protein 4 (BMP4)-based two-step protocol for differentiation of primed human pluripotent stem cells (hPSCs) into functional trophoblasts; however, those trophoblasts could not be maintained in a self-renewing TSC-like state. Here, we use the first step from this protocol, followed by a switch to newly developed TSC medium, to derive bona fide TSCs. We show that these cells resemble placenta- and naive hPSC-derived TSCs, based on their transcriptome as well as their in vitro and in vivo differentiation potential. We conclude that primed hPSCs can be used to generate functional TSCs through a simple protocol, which can be applied to a widely available set of existing hPSCs, including induced pluripotent stem cells, derived from patients with known birth outcomes.
Collapse
Affiliation(s)
- Francesca Soncin
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Robert Morey
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Tony Bui
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Daniela F Requena
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Virginia Chu Cheung
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Sampada Kallol
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Ryan Kittle
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Madeline G Jackson
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Omar Farah
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jennifer Chousal
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Morgan Meads
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Donald Pizzo
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA
| | - Mariko Horii
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Kathleen M Fisch
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Mana M Parast
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
44
|
Abstract
Treatment with bone morphogenetic protein 4 (BMP4) in human primed pluripotent stem cells (PSCs) for generating trophoblast lineage cells has sparked debate that the resulting cells are closer to amnion lineage cells rather than trophoblast. This study reports that trophoblast stem-like cells (TSLCs) can be generated from human primed PSCs by a short-term treatment of BMP4 without amnion lineage marker expression. In addition, we describe that TSLCs are self-renewing in long-term culture and bipotent as they can differentiate into functional extravillous trophoblasts and syncytiotrophoblasts. We propose an alternative method to generate an available model for studying human placental development from human primed PSCs. The placenta is a transient but important multifunctional organ crucial for healthy pregnancy for both mother and fetus. Nevertheless, limited access to human placenta samples and the paucity of a proper in vitro model system have hampered our understanding of the mechanisms underlying early human placental development and placenta-associated pregnancy complications. To overcome these constraints, we established a simple procedure with a short-term treatment of bone morphogenetic protein 4 (BMP4) in trophoblast stem cell culture medium (TSCM) to convert human primed pluripotent stem cells (PSCs) to trophoblast stem-like cells (TSLCs). These TSLCs show not only morphology and global gene expression profiles comparable to bona fide human trophoblast stem cells (TSCs) but also long-term self-renewal capacity with bipotency that allows the cells to differentiate into functional extravillous trophoblasts (EVT) and syncytiotrophoblasts (ST). These indicate that TSLCs are equivalent to genuine human TSCs. Our data suggest a straightforward approach to make human TSCs directly from preexisting primed PSCs and provide a valuable opportunity to study human placenta development and pathology from patients with placenta-related diseases.
Collapse
|
45
|
Seetharam AS, Vu HTH, Choi S, Khan T, Sheridan MA, Ezashi T, Roberts RM, Tuteja G. The product of BMP-directed differentiation protocols for human primed pluripotent stem cells is placental trophoblast and not amnion. Stem Cell Reports 2022; 17:1289-1302. [PMID: 35594861 PMCID: PMC9214062 DOI: 10.1016/j.stemcr.2022.04.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 03/24/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
The observation that trophoblast (TB) can be generated from primed pluripotent stem cells (PSCs) by exposure to bone morphogenetic protein-4 (BMP4) when FGF2 and ACTIVIN signaling is minimized has recently been challenged with the suggestion that the procedure instead produces amnion. Here, by analyzing transcriptome data from multiple sources, including bulk and single-cell data, we show that the BMP4 procedure generates bona fide TB with similarities to both placental villous TB and TB generated from TB stem cells. The analyses also suggest that the transcriptomic signatures between embryonic amnion and different forms of TB have commonalities. Our data provide justification for the continued use of TB derived from PSCs as a model for investigating placental development. Cells differentiated by using BAP protocols resemble TB more than embryonic amnion Deviation from the standard BAP protocol results in less differentiated TB Single-cell/nucleus RNA-seq analysis identifies two syncytiotrophoblast populations
Collapse
Affiliation(s)
- Arun S Seetharam
- Department of Ecology, Evolution and Organismal Biology, Iowa State University, Ames, IA, USA; Genetics Development and Cell Biology, Iowa State University, Ames, IA, USA
| | - Ha T H Vu
- Genetics Development and Cell Biology, Iowa State University, Ames, IA, USA; Bioinformatics and Computational Biology, Iowa State University, Ames, IA, USA
| | - Sehee Choi
- Christopher S Bond Life Sciences Center, University of Missouri, Columbia, MO, USA; Department of Obstetrics and Gynecology, University of Missouri School of Medicine, Columbia, MO, USA
| | - Teka Khan
- Christopher S Bond Life Sciences Center, University of Missouri, Columbia, MO, USA; Division of Animal Sciences, Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Megan A Sheridan
- Christopher S Bond Life Sciences Center, University of Missouri, Columbia, MO, USA; Department of Obstetrics and Gynecology, University of Missouri School of Medicine, Columbia, MO, USA
| | - Toshihiko Ezashi
- Christopher S Bond Life Sciences Center, University of Missouri, Columbia, MO, USA; Division of Animal Sciences, Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - R Michael Roberts
- Christopher S Bond Life Sciences Center, University of Missouri, Columbia, MO, USA; Division of Animal Sciences, Bond Life Sciences Center, University of Missouri, Columbia, MO, USA; Department of Biochemistry, University of Missouri, Columbia, MO, USA.
| | - Geetu Tuteja
- Genetics Development and Cell Biology, Iowa State University, Ames, IA, USA; Bioinformatics and Computational Biology, Iowa State University, Ames, IA, USA.
| |
Collapse
|
46
|
Physiological Function of the Dynamic Oxygen Signaling Pathway at the Maternal-fetal Interface. J Reprod Immunol 2022; 151:103626. [DOI: 10.1016/j.jri.2022.103626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 02/21/2022] [Accepted: 04/13/2022] [Indexed: 11/21/2022]
|
47
|
Functional antagonism between ΔNp63α and GCM1 regulates human trophoblast stemness and differentiation. Nat Commun 2022; 13:1626. [PMID: 35338152 PMCID: PMC8956607 DOI: 10.1038/s41467-022-29312-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/03/2022] [Indexed: 02/08/2023] Open
Abstract
The combination of EGF, CHIR99021, A83-01, SB431542, VPA, and Y27632 (EGF/CASVY) facilitates the derivation of trophoblast stem (TS) cells from human blastocysts and first-trimester, but not term, cytotrophoblasts. The mechanism underlying this chemical induction of TS cells remains elusive. Here we demonstrate that the induction efficiency of cytotrophoblast is determined by functional antagonism of the placental transcription factor GCM1 and the stemness regulator ΔNp63α. ΔNp63α reduces GCM1 transcriptional activity, whereas GCM1 inhibits ΔNp63α oligomerization and autoregulation. EGF/CASVY cocktail activates ΔNp63α, thereby partially inhibiting GCM1 activity and reverting term cytotrophoblasts into stem cells. By applying hypoxia condition, we can further reduce GCM1 activity and successfully induce term cytotrophoblasts into TS cells. Consequently, we identify mitochondrial creatine kinase 1 (CKMT1) as a key GCM1 target crucial for syncytiotrophoblast differentiation and reveal decreased CKMT1 expression in preeclampsia. Our study delineates the molecular underpinnings of trophoblast stemness and differentiation and an efficient method to establish TS cells from term placentas. Trophoblast stem cells can be derived from human blastocysts and first-trimester, but not term, cytotrophoblasts. Here the authors show that induction efficiency of cytotrophoblast is determined by antagonism between GCM1 and ΔNp63α and manipulating this antagonism facilitates derivation of TS cells from term placenta.
Collapse
|
48
|
Cui K, Zhu Y, Shi Y, Chen T, Wang H, Guo Y, Deng P, Liu H, Shao X, Qin J. Establishment of Trophoblast-Like Tissue Model from Human Pluripotent Stem Cells in Three-Dimensional Culture System. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2100031. [PMID: 34813178 PMCID: PMC8787386 DOI: 10.1002/advs.202100031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 09/23/2021] [Indexed: 06/13/2023]
Abstract
The placenta has a lifelong impact on the health of both the mother and fetus. Despite its significance, human early placental development is poorly understood due to the limited models. The models that can reflect the key features of early human placental development, especially at early gestation, are still lacking. Here, the authors report the generation of trophoblast-like tissue model from human pluripotent stem cells (hPSCs) in three-dimensional (3D) cultures. hPSCs efficiently self-organize into blastocoel-like cavities under defined conditions, which produce different trophoblast subtypes, including cytotrophoblasts (CTBs), syncytiotrophoblasts (STBs), and invasive extravillous trophoblasts (EVTs). The 3D cultures can exhibit microvilli structure and secrete human placenta-specific hormone. Single-cell RNA sequencing analysis further identifies the presence of major cell types of trophoblast-like tissue as existing in vivo. The results reveal the feasibility to establish 3D trophoblast-like tissue model from hPSCs in vitro, which is not obtained by monolayer culture. This new model system can not only facilitate to dissect the underlying mechanisms of early human placental development, but also imply its potential for study in developmental biology and gestational disorders.
Collapse
Affiliation(s)
- Kangli Cui
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yujuan Zhu
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yang Shi
- Dalian Municipal Women and Children's Medical CenterDalian116037China
| | - Tingwei Chen
- Yunnan Key Laboratory of Primate Biomedical ResearchInstitute of Primate Translational MedicineKunming University of Science and TechnologyKunming650031China
| | - Hui Wang
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yaqiong Guo
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Pengwei Deng
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Haitao Liu
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
| | - Xiaoguang Shao
- Dalian Municipal Women and Children's Medical CenterDalian116037China
| | - Jianhua Qin
- Division of BiotechnologyDalian Institute of Chemical PhysicsChinese Academy of SciencesDalian116023China
- University of Chinese Academy of SciencesBeijing100049China
- Institute for Stem Cell and RegenerationChinese Academy of SciencesBeijing100101China
- CAS Center for Excellence in Brain Science and Intelligence TechnologyChinese Academy of SciencesShanghai200031China
| |
Collapse
|
49
|
Sheridan MA, Zhao X, Fernando RC, Gardner L, Perez-Garcia V, Li Q, Marsh SGE, Hamilton R, Moffett A, Turco MY. Characterization of primary models of human trophoblast. Development 2021; 148:272500. [PMID: 34651188 PMCID: PMC8602945 DOI: 10.1242/dev.199749] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 10/01/2021] [Indexed: 01/01/2023]
Abstract
Two recently developed models, trophoblast organoids and trophoblast stem cells (TSCs), are useful tools to further the understanding of human placental development. Both differentiate from villous cytotrophoblast (VCT) to either extravillous trophoblast (EVT) or syncytiotrophoblast (SCT). Here, we compare the transcriptomes and miRNA profiles of these models to identify which trophoblast they resemble in vivo. Our findings indicate that TSCs do not readily undergo SCT differentiation and closely resemble cells at the base of the cell columns from where EVT derives. In contrast, organoids are similar to VCT and undergo spontaneous SCT differentiation. A defining feature of human trophoblast is that VCT and SCT are human leukocyte antigen (HLA) null, whereas EVT expresses HLA-C, -G and -E molecules. We find that trophoblast organoids retain these in vivo characteristics. In contrast, TSCs express classical HLA-A and HLA-B molecules, and maintain their expression after EVT differentiation, with upregulation of HLA-G. Furthermore, HLA expression in TSCs differs when grown in 3D rather than in 2D, suggesting that mechanical cues are important. Our results can be used to select the most suitable model for the study of trophoblast development, function and pathology. Summary: Characterization of trophoblast organoids and trophoblast stem cells as exciting models of human placentation enables the selection of the most suitable system to address specific research questions.
Collapse
Affiliation(s)
- Megan A Sheridan
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK.,Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Xiaohui Zhao
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK.,Department of Physiology, Neuroscience and Development, University of Cambridge, Cambridge CB2 3EG, UK
| | - Ridma C Fernando
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK.,Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Lucy Gardner
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK.,Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Vicente Perez-Garcia
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK.,Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK.,Centro de Investigación Príncipe Felipe, Eduardo Primo Yúfera, Valencia 46012, Spain
| | - Qian Li
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK.,Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | - Steven G E Marsh
- Anthony Nolan Research Institute, Royal Free Hospital, London NW3 2QG, UK.,UCL Cancer Institute, Royal Free Campus, London WC1E 6DD, UK
| | - Russell Hamilton
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK.,Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | - Ashley Moffett
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK.,Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Margherita Y Turco
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK.,Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| |
Collapse
|
50
|
Chhabra S, Warmflash A. BMP-treated human embryonic stem cells transcriptionally resemble amnion cells in the monkey embryo. Biol Open 2021; 10:271874. [PMID: 34435204 PMCID: PMC8502258 DOI: 10.1242/bio.058617] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 08/05/2021] [Indexed: 12/17/2022] Open
Abstract
Human embryonic stem cells (hESCs) possess an immense potential to generate clinically relevant cell types and unveil mechanisms underlying early human development. However, using hESCs for discovery or translation requires accurately identifying differentiated cell types through comparison with their in vivo counterparts. Here, we set out to determine the identity of much debated BMP-treated hESCs by comparing their transcriptome to recently published single cell transcriptomic data from early human embryos (
Xiang et al., 2020). Our analyses reveal several discrepancies in the published human embryo dataset, including misclassification of putative amnion, intermediate and inner cell mass cells. These misclassifications primarily resulted from similarities in pseudogene expression, highlighting the need to carefully consider gene lists when making comparisons between cell types. In the absence of a relevant human dataset, we utilized the recently published single cell transcriptome of the early post implantation monkey embryo to discern the identity of BMP-treated hESCs. Our results suggest that BMP-treated hESCs are transcriptionally more similar to amnion cells than trophectoderm cells in the monkey embryo. Together with prior studies, this result indicates that hESCs possess a unique ability to form mature trophectoderm subtypes via an amnion-like transcriptional state. This article has an associated First Person interview with the first author of the paper. Summary: We show that BMP-treated human embryonic stem cells (hESCs) are more likely to represent an amnion rather than a trophectoderm cell type.
Collapse
Affiliation(s)
- Sapna Chhabra
- Systems Synthetic and Physical Biology graduate program, Rice University, Houston, TX 77005, USA.,Department of Biosciences, Rice University, Houston, TX 77005, USA
| | - Aryeh Warmflash
- Department of Biosciences, Rice University, Houston, TX 77005, USA.,Department of Bioengineering, Rice University, Houston, TX 77005, USA
| |
Collapse
|