1
|
Pei J, Liao Y, Bai X, Li M, Wang J, Li X, Zhang H, Sui L, Kong Y. Dysregulated GLUT1 results in the pathogenesis of preeclampsia by impairing the function of trophoblast cells. Sci Rep 2024; 14:23761. [PMID: 39390043 PMCID: PMC11467397 DOI: 10.1038/s41598-024-74489-z] [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: 05/24/2024] [Accepted: 09/26/2024] [Indexed: 10/12/2024] Open
Abstract
Preeclampsia (PE) is a common placental-origin complication of pregnancy and a major cause of morbidity and mortality among pregnant women and fetuses. However, its pathogenesis has not been elucidated. Effective strategies for prevention, screening, and treatment are still lacking. Studies have indicated that dysfunction of placental trophoblast cells, such as impaired syncytialization, proliferation, and epithelial-mesenchymal transition processes, plays a crucial role in the development of PE. Glucose transporter 1 (GLUT1) is a key protein regulating glucose transport in placental tissues. However, the effect of GLUT1 on the function of trophoblast cells in PE is not well understood. In this study, we found that GLUT1 expression is reduced in PE placental tissues. GLUT1 promotes the syncytialization process by increasing the glucose uptake ability of BeWo cells. Additionally, GLUT1 promotes the proliferation, migration, and invasion capabilities of HTR-8/SVneo cells by regulating MAPK and PI3K/AKT signaling pathways. Overall, these findings provide a new insight into understanding the biological functions of GLUT1, clarifying the pathogenesis of PE, and identifying diagnostic and therapeutic targets for PE.
Collapse
Affiliation(s)
- Jingyuan Pei
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian, 116044, Liaoning Province, China
| | - Yangyou Liao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian, 116044, Liaoning Province, China
| | - Xiaoxian Bai
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian, 116044, Liaoning Province, China
| | - Min Li
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian, 116044, Liaoning Province, China
| | - Jing Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian, 116044, Liaoning Province, China
| | - Xiaotong Li
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian, 116044, Liaoning Province, China
| | - Hongshuo Zhang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Linlin Sui
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian, 116044, Liaoning Province, China.
| | - Ying Kong
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Dalian Medical University, 9 West Section, Lvshun South Road, Dalian, 116044, Liaoning Province, China.
| |
Collapse
|
2
|
Huang H, Gao S, Bao M. Exploring Mechanical Forces Shaping Self-Organization and Morphogenesis During Early Embryo Development. Annu Rev Cell Dev Biol 2024; 40:75-96. [PMID: 38608312 DOI: 10.1146/annurev-cellbio-120123-105748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2024]
Abstract
Embryonic development is a dynamic process orchestrated by a delicate interplay of biochemical and biophysical factors. While the role of genetics and biochemistry in embryogenesis has been extensively studied, recent research has highlighted the significance of mechanical regulation in shaping and guiding this intricate process. Here, we provide an overview of the current understanding of the mechanical regulation of embryo development. We explore how mechanical forces generated by cells and tissues play a crucial role in driving the development of different stages. We examine key morphogenetic processes such as compaction, blastocyst formation, implantation, and egg cylinder formation, and discuss the mechanical mechanisms and cues involved. By synthesizing the current body of literature, we highlight the emerging concepts and open questions in the field of mechanical regulation. We aim to provide an overview of the field, inspiring future investigations and fostering a deeper understanding of the mechanical aspects of embryo development.
Collapse
Affiliation(s)
- Hong Huang
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China;
| | - Shaorong Gao
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China;
| | - Min Bao
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China;
| |
Collapse
|
3
|
Stadtmauer DJ, Basanta Martínez S, Maziarz JD, Cole AG, Dagdas G, Smith GR, van Breukelen F, Pavličev M, Wagner GP. Cell type and cell signaling innovations underlying mammalian pregnancy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.01.591945. [PMID: 38746137 PMCID: PMC11092578 DOI: 10.1101/2024.05.01.591945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
How fetal and maternal cell types have co-evolved to enable mammalian placentation poses a unique evolutionary puzzle. Here, we present a multi-species atlas integrating single-cell transcriptomes from six species bracketing therian mammal diversity. We find that invasive trophoblasts share a gene-expression signature across eutherians, and evidence that endocrine decidual cells evolved stepwise from an immunomodulatory cell type retained in Tenrec with affinity to human decidua of menstruation. We recover evolutionary patterns in ligand-receptor signaling: fetal and maternal cells show a pronounced tendency towards disambiguation, but a predicted arms race dynamic between them is limited. We reconstruct cell communication networks of extinct mammalian ancestors, finding strong integration of fetal trophoblast into maternal networks. Together, our results reveal a dynamic history of cell type and signaling evolution. Synopsis The fetal-maternal interface is one of the most intense loci of cell-cell signaling in the human body. Invasion of cells from the fetal placenta into the uterus, and the corresponding transformation of maternal tissues called decidualization, first evolved in the stem lineage of eutherian mammals( 1 , 2 ). Single-cell studies of the human fetal-maternal interface have provided new insight into the cell type diversity and cell-cell interactions governing this chimeric organ( 3-5 ). However, the fetal-maternal interface is also one of the most rapidly evolving, and hence most diverse, characters among mammals( 6 ), and an evolutionary analysis is missing. Here, we present and compare single-cell data from the fetal-maternal interface of species bracketing key events in mammal phylogeny: a marsupial (opossum, Monodelphis domestica ), the afrotherian Tenrec ecaudatus, and four Euarchontoglires - guinea pig and mouse (Rodentia) together with recent macaque and human data (primates) ( 4 , 5 , 7 ). We infer cell type homologies, identify a gene-expression signature of eutherian invasive trophoblast conserved over 99 million years, and discover a predecidual cell in the tenrec which suggests stepwise evolution of the decidual stromal cell. We reconstruct ancestral cell signaling networks, revealing the integration of fetal cell types into the interface. Finally, we test two long-standing theoretical predictions, the disambiguation hypothesis( 8 ) and escalation hypothesis( 9 ), at transcriptome-wide scale, finding divergence between fetal and maternal signaling repertoires but arms race dynamics restricted to a small subset of ligand-receptor pairs. In so doing, we trace the co-evolutionary history of cell types and their signaling across mammalian viviparity.
Collapse
|
4
|
Pan L, Zhu F, Yu A, Jia C, Tang H, Zhou M, Li M, Jiang S, Li J, Cui Y, Tang L. Effect of bromodomain PHD-finger transcription factor (BPTF) on trophoblast epithelial-to-mesenchymal transition. Gene 2024; 914:148405. [PMID: 38521110 DOI: 10.1016/j.gene.2024.148405] [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: 12/17/2023] [Revised: 03/03/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
The trophoblast epithelial-to-mesenchymal transition (EMT) is a procedure related to embryo implantation, spiral artery establishment and fetal-maternal communication, which is a key event for successful pregnancy. Inadequate EMT is one of the pathological mechanisms of recurrent miscarriage (RM). Whole-exome sequencing revealed that the mutation of bromodomain PHD-finger transcription factor (BPTF) was strongly associated with RM. In the present study, the effects of BPTF on EMT and the underlying mechanism were investigated. We found that the expression of BPTF in the villi of RM patients was significantly downregulated. Gene Ontology (GO) analysis revealed that BPTF participated in cell adhesion. The knockdown of BPTF prevented EMT and attenuated trophoblast invasion in vitro. BPTF activated Slug transcription by binding directly to the promoter region of the Slug gene. Interestingly, the protein levels of both Slug and BPTF were decreased in the villous cytotrophoblasts (VCTs) of RM villi. In conclusion, BPTF participates in the regulation of trophoblast EMT by activating Slug expression, suggesting that BPTF defects are an important factor in RM pathogenesis.
Collapse
Affiliation(s)
- Linqing Pan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; Clinical Center of Reproductive Medicine, Lianyungang Maternal and Child Health Hospital, Kangda College of Nanjing Medical University, Lianyungang 222000, China
| | - Fuquan Zhu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Aochen Yu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Chao Jia
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Huaiyun Tang
- Clinical Center of Reproductive Medicine, Lianyungang Maternal and Child Health Hospital, Kangda College of Nanjing Medical University, Lianyungang 222000, China
| | - Minglian Zhou
- Clinical Center of Reproductive Medicine, Lianyungang Maternal and Child Health Hospital, Kangda College of Nanjing Medical University, Lianyungang 222000, China
| | - Mingrui Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center of Clinical Reproductive Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Shiwen Jiang
- Clinical Center of Reproductive Medicine, Lianyungang Maternal and Child Health Hospital, Kangda College of Nanjing Medical University, Lianyungang 222000, China
| | - Juan Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Yugui Cui
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center of Clinical Reproductive Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| | - Lisha Tang
- Clinical Center of Reproductive Medicine, Lianyungang Maternal and Child Health Hospital, Kangda College of Nanjing Medical University, Lianyungang 222000, China.
| |
Collapse
|
5
|
Chen Y, Ye X, Zhong Y, Kang X, Tang Y, Zhu H, Pang C, Ning S, Liang S, Zhang F, Li C, Li J, Gu C, Cheng Y, Kuang Z, Qiu J, Jin J, Luo H, Fu M, Hui HX, Li L, Ruan D, Liu P, Chen X, Sun L, Ai S, Gao X. SP6 controls human cytotrophoblast fate decisions and trophoblast stem cell establishment by targeting MSX2 regulatory elements. Dev Cell 2024; 59:1506-1522.e11. [PMID: 38582082 DOI: 10.1016/j.devcel.2024.03.025] [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: 07/27/2023] [Revised: 12/14/2023] [Accepted: 03/12/2024] [Indexed: 04/08/2024]
Abstract
The commitment and differentiation of human placental progenitor cytotrophoblast (CT) cells are crucial for a successful pregnancy, but the underlying mechanism remains poorly understood. Here, we identified the transcription factor (TF), specificity protein 6 (SP6), as a human species-specific trophoblast lineage TF expressed in human placental CT cells. Using pluripotent stem cells as a model, we demonstrated that SP6 controls CT generation and the establishment of trophoblast stem cells (TSCs) and identified msh homeobox 2 (MSX2) as the downstream effector in these events. Mechanistically, we showed that SP6 interacts with histone acetyltransferase P300 to alter the landscape of H3K27ac at targeted regulatory elements, thereby favoring transcriptional activation and facilitating CT cell fate decisions and TSC maintenance. Our results established SP6 as a regulator of the human trophoblast lineage and implied its role in placental development and the pathogenies of placental diseases.
Collapse
Affiliation(s)
- Yanglin Chen
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xianhua Ye
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yulong Zhong
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiangjin Kang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Yanqing Tang
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Haoyun Zhu
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Changmiao Pang
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shaoqiang Ning
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shiqing Liang
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Feifan Zhang
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Chao Li
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jie Li
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Chengtao Gu
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yuanxiong Cheng
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, Guangdong, China
| | - Zhanpeng Kuang
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jingyang Qiu
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jin Jin
- Department of Gynaecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Haisi Luo
- Department of Gynaecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Mingyu Fu
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hannah Xiaoyan Hui
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Lei Li
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China; Key Laboratory for Reproductive Medicine of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510515, China
| | - Degong Ruan
- School of Biomedical Sciences, Stem Cell, and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Pentao Liu
- School of Biomedical Sciences, Stem Cell, and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Xi Chen
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Liangzhong Sun
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Shanshan Ai
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Xuefei Gao
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, Guangdong, China; Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
6
|
Yu D, Wan H, Tong C, Guang L, Chen G, Su J, Zhang L, Wang Y, Xiao Z, Zhai J, Yan L, Ma W, Liang K, Liu T, Wang Y, Peng Z, Luo L, Yu R, Li W, Qi H, Wang H, Shyh-Chang N. A multi-tissue metabolome atlas of primate pregnancy. Cell 2024; 187:764-781.e14. [PMID: 38306985 DOI: 10.1016/j.cell.2023.11.043] [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: 11/08/2022] [Revised: 08/08/2023] [Accepted: 11/29/2023] [Indexed: 02/04/2024]
Abstract
Pregnancy induces dramatic metabolic changes in females; yet, the intricacies of this metabolic reprogramming remain poorly understood, especially in primates. Using cynomolgus monkeys, we constructed a comprehensive multi-tissue metabolome atlas, analyzing 273 samples from 23 maternal tissues during pregnancy. We discovered a decline in metabolic coupling between tissues as pregnancy progressed. Core metabolic pathways that were rewired during primate pregnancy included steroidogenesis, fatty acid metabolism, and arachidonic acid metabolism. Our atlas revealed 91 pregnancy-adaptive metabolites changing consistently across 23 tissues, whose roles we verified in human cell models and patient samples. Corticosterone and palmitoyl-carnitine regulated placental maturation and maternal tissue progenitors, respectively, with implications for maternal preeclampsia, diabetes, cardiac hypertrophy, and muscle and liver regeneration. Moreover, we found that corticosterone deficiency induced preeclampsia-like inflammation, indicating the atlas's potential clinical value. Overall, our multi-tissue metabolome atlas serves as a framework for elucidating the role of metabolic regulation in female health during pregnancy.
Collapse
Affiliation(s)
- Dainan Yu
- Key Laboratory of Organ Regeneration and Reconstruction, 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; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Haifeng Wan
- Key Laboratory of Organ Regeneration and Reconstruction, 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; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Chao Tong
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Lu Guang
- Key Laboratory of Organ Regeneration and Reconstruction, 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; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Gang Chen
- Key Laboratory of Organ Regeneration and Reconstruction, 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; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Jiali Su
- Key Laboratory of Organ Regeneration and Reconstruction, 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; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Lan Zhang
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yue Wang
- Key Laboratory of Organ Regeneration and Reconstruction, 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; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Zhenyu Xiao
- Key Laboratory of Organ Regeneration and Reconstruction, 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; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Jinglei Zhai
- Key Laboratory of Organ Regeneration and Reconstruction, 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; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Long Yan
- Key Laboratory of Organ Regeneration and Reconstruction, 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; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Wenwu Ma
- Key Laboratory of Organ Regeneration and Reconstruction, 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; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Kun Liang
- Key Laboratory of Organ Regeneration and Reconstruction, 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; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Taoyan Liu
- Key Laboratory of Organ Regeneration and Reconstruction, 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; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yuefan Wang
- Key Laboratory of Organ Regeneration and Reconstruction, 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; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Zehang Peng
- Key Laboratory of Organ Regeneration and Reconstruction, 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; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Lanfang Luo
- Key Laboratory of Organ Regeneration and Reconstruction, 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; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Ruoxuan Yu
- Key Laboratory of Organ Regeneration and Reconstruction, 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; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Wei Li
- Key Laboratory of Organ Regeneration and Reconstruction, 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; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China.
| | - Hongbo Qi
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing 401120, China.
| | - Hongmei Wang
- Key Laboratory of Organ Regeneration and Reconstruction, 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; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China.
| | - Ng Shyh-Chang
- Key Laboratory of Organ Regeneration and Reconstruction, 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; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China.
| |
Collapse
|
7
|
Yoshida K, Kusama K, Shinohara G, Sato S, Yoshie M, Tamura K. Quercetin stimulates trophoblast fusion via the mitochondrial function. Sci Rep 2024; 14:287. [PMID: 38168580 PMCID: PMC10762005 DOI: 10.1038/s41598-023-50712-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/23/2023] [Indexed: 01/05/2024] Open
Abstract
The fusion of mononuclear trophoblasts into multinucleate syncytiotrophoblasts is the critical event in the process of syncytialization, and its dysregulation can lead to pregnancy complications, notably hypertensive disorders of pregnancy (HDP). Oxidative stress may disrupt trophoblast syncytialization in HDP. Specifically, placentas with HDP exhibit impaired mitochondria, giving rise to the generation of reactive oxygen species (ROS) and subsequent oxidative stress. Quercetin, a bioflavonoid known for its antioxidant and anti-aging properties, has the potential to mitigate oxidative stress during trophoblast syncytialization. However, the precise mechanism underlying the action of quercetin in these processes remains to be elucidated. To explore the impact of quercetin on syncytialization, mitochondrial function, and ROS generation, cyclic AMP-stimulated BeWo cells were treated with quercetin. The expression of markers associated with cell fusion, mitochondrial function, and oxidative stress was determined using qPCR and western blotting. Additionally, morphological syncytialization and mitophagy (mitochondrial degradation) were assessed by immunofluorescence analysis. Our results revealed that quercetin increased the expression of syncytialization markers and promoted cell fusion. Furthermore, this compound also upregulated markers associated with mitophagy and mitochondrial fusion, which are corroborated by visual evidence of mitophagy through the fluorescence microscope. Cell fusion naturally stimulated ROS generation, which was attenuated by quercetin. Quercetin downregulated the expression of NRF2 and HO-1 during syncytialization, while increasing the expression of sirtuin1/3/6, which are known to play essential roles in antioxidant responses. In conclusion, quercetin effectively regulates mitochondrial function through its antioxidant properties and the suppression of ROS generation, ultimately promoting trophoblast fusion, suggesting that the flavonoid has the potential to ameliorate pregnancy-related disorder stemming from placental dysplasia.
Collapse
Affiliation(s)
- Kanoko Yoshida
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Kazuya Kusama
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan.
| | - Go Shinohara
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Shiho Sato
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Mikihiro Yoshie
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Kazuhiro Tamura
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| |
Collapse
|
8
|
Yuan X, Liu X, Zhu F, Huang B, Lin L, Huang J, Wen L, Kilby MD, Baker PN, Fu Y, Wu W, Qi H, Tang J, Tong C. Endoplasmic reticulum stress impairs trophoblast syncytialization through upregulation of HtrA4 and causes early-onset preeclampsia. J Hypertens 2023; 41:2095-2106. [PMID: 37728094 DOI: 10.1097/hjh.0000000000003541] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023]
Abstract
OBJECTIVE Syncytiotrophoblasts form via mononuclear cytotrophoblast fusion during placentation and play a critical role in maternal-fetal communication. Impaired syncytialization inevitably leads to pregnancy-associated complications, including preeclampsia. Endoplasmic reticulum stress (ERS) is reportedly linked with preeclampsia, but little is known about its association with syncytialization. High temperature requirement factor A4 (HtrA4), a placental-specific protease, is responsible for protein quality control and placental syncytialization. This study aimed to investigate the relationship among HtrA4, ERS, and trophoblast syncytialization in the development of early-onset preeclampsia (EO-PE). METHODS HtrA4 expression and ERS in preeclamptic placentas and control placentas were analyzed by Western blotting and qRT-PCR. HtrA4 and ERS localization in placentas was determined by immunohistochemistry and immunofluorescence. BeWo cells were used to stimulate the effects of HtrA4 and ERS on syncytialization. RESULTS HtrA4 expression was upregulated in EO-PE and positively correlated with ERS. HtrA4 activity was increased in preeclampsia. Under normoxia, HtrA4 overexpression in BeWo cells did not alter the ERS level. In addition, treatment with hypoxia/reoxygenation (H/R) or an ERS inducer increased HtrA4 expression. HtrA4 upregulation suppressed the levels of syncytin-2 and β-HCG in the presence of forskolin (FSK), and this change was exaggerated after ERS activation. In addition, treatment with an ERS inhibitor markedly suppressed FSK-treated cell fusion in a manner related to downregulation of HtrA4 expression. CONCLUSION Our results suggest that ERS enables syncytialization of placental development by upregulating HtrA4, but that excessive HtrA4 expression and preexisting ERS impair syncytialization and cause EO-PE.
Collapse
Affiliation(s)
- Xi Yuan
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University
- International Collaborative Laboratory of Reproduction and Development of the Chinese Ministry of Education, Chongqing Medical University
| | - Xiyao Liu
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University
- International Collaborative Laboratory of Reproduction and Development of the Chinese Ministry of Education, Chongqing Medical University
| | - Fangyu Zhu
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University
- International Collaborative Laboratory of Reproduction and Development of the Chinese Ministry of Education, Chongqing Medical University
| | - Biao Huang
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University
- International Collaborative Laboratory of Reproduction and Development of the Chinese Ministry of Education, Chongqing Medical University
| | - Li Lin
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University
- International Collaborative Laboratory of Reproduction and Development of the Chinese Ministry of Education, Chongqing Medical University
| | - Jiayu Huang
- Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | | - Mark D Kilby
- Fetal Medicine Centre, Birmingham Women's & Children's Foundation Trust
- Institute of Metabolism & Systems Research, College of Medical & Dental Sciences, University of Birmingham, Birmingham
| | - Philip N Baker
- College of Life Sciences, University of Leicester, Leicester, UK
| | - Yong Fu
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University
- International Collaborative Laboratory of Reproduction and Development of the Chinese Ministry of Education, Chongqing Medical University
| | - Weiwei Wu
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi
| | - Hongbo Qi
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University
- International Collaborative Laboratory of Reproduction and Development of the Chinese Ministry of Education, Chongqing Medical University
- Department of Obstetrics, Women and Children's Hospital of Chongqing Medical University
| | - Jing Tang
- International Collaborative Laboratory of Reproduction and Development of the Chinese Ministry of Education, Chongqing Medical University
- School of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Chao Tong
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, The First Affiliated Hospital of Chongqing Medical University
- International Collaborative Laboratory of Reproduction and Development of the Chinese Ministry of Education, Chongqing Medical University
| |
Collapse
|
9
|
Shimode S. Acquisition and Exaptation of Endogenous Retroviruses in Mammalian Placenta. Biomolecules 2023; 13:1482. [PMID: 37892164 PMCID: PMC10604696 DOI: 10.3390/biom13101482] [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: 08/15/2023] [Revised: 10/02/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2023] Open
Abstract
Endogenous retroviruses (ERVs) are retrovirus-like sequences that were previously integrated into the host genome. Although most ERVs are inactivated by mutations, deletions, or epigenetic regulation, some remain transcriptionally active and impact host physiology. Several ERV-encoded proteins, such as Syncytins and Suppressyn, contribute to placenta acquisition, a crucial adaptation in mammals that protects the fetus from external threats and other risks while enabling the maternal supply of oxygen, nutrients, and antibodies. In primates, Syncytin-1 and Syncytin-2 facilitate cell-cell fusion for placental formation. Suppressyn is the first ERV-derived protein that inhibits cell fusion by binding to ASCT2, the receptor for Syncytin-1. Furthermore, Syncytin-2 likely inserted into the genome of the common ancestor of Anthropoidea, whereas Syncytin-1 and Suppressyn likely inserted into the ancestor of catarrhines; however, they were inactivated in some lineages, suggesting that multiple exaptation events had occurred. This review discusses the role of ERV-encoded proteins, particularly Syncytins and Suppressyn, in placental development and function, focusing on the integration of ERVs into the host genome and their contribution to the genetic mechanisms underlying placentogenesis. This review provides valuable insights into the molecular and genetic aspects of placentation, potentially shedding light on broader evolutionary and physiological processes in mammals.
Collapse
Affiliation(s)
- Sayumi Shimode
- Genome Editing Innovation Center, Hiroshima University, Higashi-Hiroshima, Hiroshima 739-0046, Japan;
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima 739-8526, Japan
| |
Collapse
|
10
|
Morey R, Bui T, Fisch KM, Horii M. Modeling placental development and disease using human pluripotent stem cells. Placenta 2023; 141:18-25. [PMID: 36333266 PMCID: PMC10148925 DOI: 10.1016/j.placenta.2022.10.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/03/2022] [Accepted: 10/18/2022] [Indexed: 11/08/2022]
Abstract
Our current knowledge of the cellular and molecular mechanisms of placental epithelial cells, trophoblast, primarily came from the use of mouse trophoblast stem cells and tumor-derived or immortalized human trophoblast cell lines. This was mainly due to the difficulties in maintaining primary trophoblast in culture and establishing human trophoblast stem cell (hTSC) lines. However, in-depth characterization of these cellular models and in vivo human trophoblast have revealed significant discrepancies. For the past two decades, multiple groups have shown that human pluripotent stem cells (hPSCs) can be differentiated into trophoblast, and thus could be used as a model for normal and disease trophoblast differentiation. During this time, trophoblast differentiation protocols have evolved, enabling researchers to study cellular characteristics at trophectoderm (TE), trophoblast stem cells (TSC), syncytiotrophoblast (STB), and extravillous trophoblast (EVT) stages. Recently, several groups reported methods to derive hTSC from pre-implantation blastocyst or early gestation placenta, and trophoblast organoids from early gestation placenta, drastically changing the landscape of trophoblast research. These culture conditions have been rapidly applied to generate hPSC-derived TSC and trophoblast organoids. As a result of these technological advancements, the field's capacity to better understand trophoblast differentiation and their involvement in pregnancy related disease has greatly expanded. Here, we present in vitro models of human trophoblast differentiation, describing both primary and hPSC-derived TSC, maintained as monolayers and 3-dimensional trophoblast organoids, as a tool to study early placental development and disease in multiple settings.
Collapse
Affiliation(s)
- Robert Morey
- Department of Pathology, University of California San Diego, La Jolla, CA, 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Tony Bui
- Department of Pathology, University of California San Diego, La Jolla, CA, 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Kathleen M Fisch
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Mariko Horii
- Department of Pathology, University of California San Diego, La Jolla, CA, 92093, USA; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
11
|
Bueno-Sánchez JC, Gómez-Gutiérrez AM, Maldonado-Estrada JG, Quintana-Castillo JC. Expression of placental glycans and its role in regulating peripheral blood NK cells during preeclampsia: a perspective. Front Endocrinol (Lausanne) 2023; 14:1087845. [PMID: 37206444 PMCID: PMC10190602 DOI: 10.3389/fendo.2023.1087845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 03/03/2023] [Indexed: 05/21/2023] Open
Abstract
Preeclampsia is a pregnancy-related multisystem disorder characterized by altered trophoblast invasion, oxidative stress, exacerbation of systemic inflammatory response, and endothelial damage. The pathogenesis includes hypertension and mild-to-severe microangiopathy in the kidney, liver, placenta, and brain. The main mechanisms involved in its pathogenesis have been proposed to limit trophoblast invasion and increase the release of extracellular vesicles from the syncytiotrophoblast into the maternal circulation, exacerbating the systemic inflammatory response. The placenta expresses glycans as part of its development and maternal immune tolerance during gestation. The expression profile of glycans at the maternal-fetal interface may play a fundamental role in physiological pregnancy changes and disorders such as preeclampsia. It is unclear whether glycans and their lectin-like receptors are involved in the mechanisms of maternal-fetal recognition by immune cells during pregnancy homeostasis. The expression profile of glycans appears to be altered in hypertensive disorders of pregnancy, which could lead to alterations in the placental microenvironment and vascular endothelium in pregnancy conditions such as preeclampsia. Glycans with immunomodulatory properties at the maternal-fetal interface are altered in early-onset severe preeclampsia, implying that innate immune system components, such as NK cells, exacerbate the systemic inflammatory response observed in preeclampsia. In this article, we discuss the evidence for the role of glycans in gestational physiology and the perspective of glycobiology on the pathophysiology of hypertensive disorders in gestation.
Collapse
Affiliation(s)
- Julio C. Bueno-Sánchez
- Reproduction Group, Department of Physiology and Biochemistry, School of Medicine, Universidad de Antioquia, Medellín, Colombia
- Department of Obstetrics and Gynecology, School of Medicine, Universidad de Antioquia, Medellín, Colombia
- Red Iberoamericana de Alteraciones Vasculares en Trastornos del Embarazo (RIVATREM), Chillan, Chile
| | - Alejandra M. Gómez-Gutiérrez
- Reproduction Group, Department of Physiology and Biochemistry, School of Medicine, Universidad de Antioquia, Medellín, Colombia
| | - Juan G. Maldonado-Estrada
- One Health and Veterinary Innovative Research & Development (OHVRI) Research Group, Escuela de Medicina Veterinaria, Universidad de Antioquia, Medellín, Colombia
| | | |
Collapse
|
12
|
Mohammad S, Bhattacharjee J, Tzaneva V, Hutchinson KA, Shaikh M, Fernandes da Silva D, Burger D, Adamo KB. The Influence of Exercise-Associated Small Extracellular Vesicles on Trophoblasts In Vitro. Biomedicines 2023; 11:biomedicines11030857. [PMID: 36979835 PMCID: PMC10045992 DOI: 10.3390/biomedicines11030857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/01/2023] [Accepted: 03/09/2023] [Indexed: 03/15/2023] Open
Abstract
Exercise induces the release of small extracellular vesicles (sEVs) into circulation that are postulated to mediate tissue cross-talk during exercise. We previously reported that pregnant individuals released greater levels of sEVs into circulation after exercise compared to matched non-pregnant controls, but their biological functions remain unknown. In this study, sEVs isolated from the plasma of healthy pregnant and non-pregnant participants after a single bout of moderate-intensity exercise were evaluated for their impact on trophoblasts in vitro. Exercise-associated sEVs were found localized within the cytoplasm of BeWo choriocarcinoma cells, used to model trophoblasts in vitro. Exposure to exercise-associated sEVs did not significantly alter BeWo cell proliferation, gene expression of angiogenic growth factors VEGF and PLGF, or the release of the hormone human chorionic gonadotropin. The results from this pilot study support that exercise-associated sEVs could interact with trophoblasts in vitro, and warrant further investigation to reveal their potential role in communicating the effects of exercise to the maternal–fetal interface.
Collapse
Affiliation(s)
- Shuhiba Mohammad
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Jayonta Bhattacharjee
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Velislava Tzaneva
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Kelly Ann Hutchinson
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Madeeha Shaikh
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Danilo Fernandes da Silva
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Dylan Burger
- Kidney Research Centre, Department of Cellular and Molecular Medicine, The Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON K1H 8L6, Canada
| | - Kristi B. Adamo
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
- Correspondence:
| |
Collapse
|
13
|
Krop J, Tian X, van der Hoorn ML, Eikmans M. The Mac Is Back: The Role of Macrophages in Human Healthy and Complicated Pregnancies. Int J Mol Sci 2023; 24:5300. [PMID: 36982375 PMCID: PMC10049527 DOI: 10.3390/ijms24065300] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/12/2023] Open
Abstract
Pregnancy is a fascinating immunological paradox: the semi-allogeneic fetus generally grows without any complications. In the placenta, fetal trophoblast cells come into contact with maternal immune cells. Inaccurate or inadequate adaptations of the maternal immune system could lead to problems with the functioning of the placenta. Macrophages are important for tissue homeostasis, cleanup, and the repair of damaged tissues. This is crucial for a rapidly developing organ such as the placenta. The consensus on macrophages at the maternal-fetal interface in pregnancy is that a major proportion have an anti-inflammatory, M2-like phenotype, that expresses scavenger receptors and is involved in tissue remodeling and the dampening of the immune reactions. Recent multidimensional analyses have contributed to a more detailed outlook on macrophages. The new view is that this lineage represents a highly diverse phenotype and is more prevalent than previously thought. Spatial-temporal in situ analyses during gestation have identified unique interactions of macrophages both with trophoblasts and with T cells at different trimesters of pregnancy. Here, we elaborate on the role of macrophages during early human pregnancy and at later gestation. Their possible effect is reviewed in the context of HLA incompatibility between mother and fetus, first in naturally conceived pregnancies, but foremost in pregnancies after oocyte donation. The potential functional consequences of macrophages for pregnancy-related immune reactions and the outcome in patients with recurrent pregnancy loss are also discussed.
Collapse
Affiliation(s)
- Juliette Krop
- Department of Immunology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Xuezi Tian
- Department of Immunology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Department of Obstetrics and Gynaecology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Marie-Louise van der Hoorn
- Department of Obstetrics and Gynaecology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Michael Eikmans
- Department of Immunology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
14
|
Zorzan I, Betto RM, Rossignoli G, Arboit M, Drusin A, Corridori C, Martini P, Martello G. Chemical conversion of human conventional PSCs to TSCs following transient naive gene activation. EMBO Rep 2023; 24:e55235. [PMID: 36847616 PMCID: PMC10074076 DOI: 10.15252/embr.202255235] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 01/31/2023] [Accepted: 02/03/2023] [Indexed: 03/01/2023] Open
Abstract
In human embryos, naive pluripotent cells of the inner cell mass (ICM) generate epiblast, primitive endoderm and trophectoderm (TE) lineages, whence trophoblast cells derive. In vitro, naive pluripotent stem cells (PSCs) retain this potential and efficiently generate trophoblast stem cells (TSCs), while conventional PSCs form TSCs at low efficiency. Transient histone deacetylase and MEK inhibition combined with LIF stimulation is used to chemically reset conventional to naive PSCs. Here, we report that chemical resetting induces the expression of both naive and TSC markers and of placental imprinted genes. A modified chemical resetting protocol allows for the fast and efficient conversion of conventional PSCs into TSCs, entailing shutdown of pluripotency genes and full activation of the trophoblast master regulators, without induction of amnion markers. Chemical resetting generates a plastic intermediate state, characterised by co-expression of naive and TSC markers, after which cells steer towards one of the two fates in response to the signalling environment. The efficiency and rapidity of our system will be useful to study cell fate transitions and to generate models of placental disorders.
Collapse
Affiliation(s)
- Irene Zorzan
- Department of Molecular Medicine, Medical School, University of Padua, Padua, Italy
| | | | | | - Mattia Arboit
- Department of Biology, University of Padua, Padua, Italy
| | - Andrea Drusin
- Department of Biology, University of Padua, Padua, Italy
| | | | - Paolo Martini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | |
Collapse
|
15
|
Cáceres D, Ochoa M, González-Ortiz M, Bravo K, Eugenín J. Effects of Prenatal Cannabinoids Exposure upon Placenta and Development of Respiratory Neural Circuits. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1428:199-232. [PMID: 37466775 DOI: 10.1007/978-3-031-32554-0_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Cannabis use has risen dangerously during pregnancy in the face of incipient therapeutic use and a growing perception of safety. The main psychoactive compound of the Cannabis sativa plant is the phytocannabinoid delta-9-tetrahydrocannabinol (A-9 THC), and its status as a teratogen is controversial. THC and its endogenous analogues, anandamide (AEA) and 2-AG, exert their actions through specific receptors (eCBr) that activate intracellular signaling pathways. CB1r and CB2r, also called classic cannabinoid receptors, together with their endogenous ligands and the enzymes that synthesize and degrade them, constitute the endocannabinoid system. This system is distributed ubiquitously in various central and peripheral tissues. Although the endocannabinoid system's most studied role is controlling the release of neurotransmitters in the central nervous system, the study of long-term exposure to cannabinoids on fetal development is not well known and is vital for understanding environmental or pathological embryo-fetal or postnatal conditions. Prenatal exposure to cannabinoids in animal models has induced changes in placental and embryo-fetal organs. Particularly, cannabinoids could influence both neural and nonneural tissues and induce embryo-fetal pathological conditions in critical processes such as neural respiratory control. This review aims at the acute and chronic effects of prenatal exposure to cannabinoids on placental function and the embryo-fetal neurodevelopment of the respiratory pattern. The information provided here will serve as a theoretical framework to critically evaluate the teratogen effects of the consumption of cannabis during pregnancy.
Collapse
Affiliation(s)
- Daniela Cáceres
- Laboratorio de Sistemas Neurales, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Martín Ochoa
- Laboratorio de Sistemas Neurales, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Marcelo González-Ortiz
- Laboratorio de Investigación Materno-Fetal (LIMaF), Departamento de Obstetricia y Ginecología, Facultad de Medicina, Universidad de Concepción, Concepción, Chile
| | - Karina Bravo
- Laboratorio de Sistemas Neurales, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
- Facultad de Ingeniería, Universidad Autónoma de Chile, Providencia, Chile
| | - Jaime Eugenín
- Laboratorio de Sistemas Neurales, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile.
| |
Collapse
|
16
|
Matsumoto S, Okamura E, Muto M, Ema M. Similarities and differences in placental development between humans and cynomolgus monkeys. Reprod Med Biol 2023; 22:e12522. [PMID: 37377753 PMCID: PMC10292683 DOI: 10.1002/rmb2.12522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/07/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Background The placenta is an extraembryonic organ, which is essential to maintain a normal pregnancy. However, placental development in humans is poorly understood because of technical and ethical reasons. Methods We analyzed the anatomical localization of each trophoblastic subtype in the cynomolgus monkey placenta by immunohistochemistry in the early second trimester. Histological differences among the mouse, cynomolgus monkey, and human placenta were compared. The PubMed database was used to search for studies on placentation in rodents and primates. Main findings The anatomical structures and subtypes of the placenta in cynomolgus monkeys are highly similar to those in humans, with the exception of fewer interstitial extravillous trophoblasts in cynomolgus monkeys. Conclusion The cynomolgus monkey appears to be a good animal model to investigate human placentation.
Collapse
Affiliation(s)
- Shoma Matsumoto
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life ScienceShiga University of Medical ScienceOtsuJapan
| | - Eiichi Okamura
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life ScienceShiga University of Medical ScienceOtsuJapan
| | - Masanaga Muto
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life ScienceShiga University of Medical ScienceOtsuJapan
| | - Masatsugu Ema
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life ScienceShiga University of Medical ScienceOtsuJapan
- Institute for the Advanced Study of Human Biology (WPI‐ASHBi)Kyoto UniversityKyotoJapan
| |
Collapse
|
17
|
Mizutani T, Orisaka M, Miyazaki Y, Morichika R, Uesaka M, Miyamoto K, Yoshida Y. Inhibition of YAP/TAZ-TEAD activity induces cytotrophoblast differentiation into syncytiotrophoblast in human trophoblast. Mol Hum Reprod 2022; 28:6673154. [PMID: 35993908 DOI: 10.1093/molehr/gaac032] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
During placentation, placental cytotrophoblast (CT) cells differentiate into syncytiotrophoblast (ST) cells and extravillous trophoblast (EVT) cells. In the placenta, the expression of various genes is regulated by the Hippo pathway through a transcription complex, Yes-associated protein (YAP)/transcriptional coactivator with PDZ-binding motif (TAZ)-TEA domain transcription factor (TEAD) (YAP/TAZ-TEAD) activity. YAP/TAZ-TEAD activity is controlled by multiple factors and signaling, such as cyclic AMP (cAMP) signaling. cAMP signaling is believed to be involved in the regulation of trophoblast function but is not yet fully understood. Here we showed that YAP/TAZ-TEAD expressions and their activities were altered by cAMP stimulation in BeWo cells, a human choriocarcinoma cell line. The repression of YAP/TAZ-TEAD activity induced the expression of ST-specific genes without cAMP stimulation, and transduction of constitutively active YAP, i.e., YAP-5SA, resulted in the repression of 8Br-cAMP-induced expressions of ST-specific genes in a TEAD-dependent manner. We also investigated the role of YAP/TAZ-TEAD in maintaining CT cells and their differentiation into ST and EVT cells using human trophoblast stem (TS) cells. YAP/TAZ-TEAD activity was involved in maintaining the stemness of TS cells. Induction or repression of YAP/TAZ-TEAD activity resulted in marked changes in the expression of ST-specific genes. Using primary CT cells, which spontaneously differentiate into ST-like cells, the effects of YAP-5SA transduction were investigated, and the expression of ST-specific genes was found to be repressed. These results indicate that the inhibition of YAP/TAZ-TEAD activity, with or without cAMP stimulation, is essential for the differentiation of CT cells into ST cells.
Collapse
Affiliation(s)
- Tetsuya Mizutani
- Department of Nursing, Faculty of Nursing and Welfare Sciences, Fukui Prefectural University, Japan
| | - Makoto Orisaka
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Japan
| | - Yumiko Miyazaki
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Japan
| | - Ririko Morichika
- Department of Nursing, Faculty of Nursing and Welfare Sciences, Fukui Prefectural University, Japan
| | - Miki Uesaka
- Department of Nursing, Faculty of Nursing and Welfare Sciences, Fukui Prefectural University, Japan.,Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Japan
| | | | - Yoshio Yoshida
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Japan
| |
Collapse
|
18
|
Imakawa K, Kusama K, Kaneko-Ishino T, Nakagawa S, Kitao K, Miyazawa T, Ishino F. Endogenous Retroviruses and Placental Evolution, Development, and Diversity. Cells 2022; 11:cells11152458. [PMID: 35954303 PMCID: PMC9367772 DOI: 10.3390/cells11152458] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/01/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022] Open
Abstract
The main roles of placentas include physical protection, nutrient and oxygen import, export of gasses and fetal waste products, and endocrinological regulation. In addition to physical protection of the fetus, the placentas must provide immune protection throughout gestation. These basic functions are well-conserved; however, placentas are undoubtedly recent evolving organs with structural and cellular diversities. These differences have been explained for the last two decades through co-opting genes and gene control elements derived from transposable elements, including endogenous retroviruses (ERVs). However, the differences in placental structures have not been explained or characterized. This manuscript addresses the sorting of ERVs and their integration into the mammalian genomes and provides new ways to explain why placental structures have diverged.
Collapse
Affiliation(s)
- Kazuhiko Imakawa
- Research Institute of Agriculture, Tokai University, Kumamoto 862-8652, Japan
- Correspondence: ; Tel.: +81-96-386-2652
| | - Kazuya Kusama
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | | | - So Nakagawa
- Department of Molecular Life Science, Tokai University School of Medicine, Nakagawa 259-1193, Japan
| | - Koichi Kitao
- Laboratory of Virus-Host Coevolution, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Takayuki Miyazawa
- Laboratory of Virus-Host Coevolution, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Fumitoshi Ishino
- Institute of Research, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| |
Collapse
|
19
|
Chen Y, Siriwardena D, Penfold C, Pavlinek A, Boroviak TE. An integrated atlas of human placental development delineates essential regulators of trophoblast stem cells. Development 2022; 149:275917. [PMID: 35792865 PMCID: PMC9340556 DOI: 10.1242/dev.200171] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 05/12/2022] [Indexed: 12/21/2022]
Abstract
The trophoblast lineage safeguards fetal development by mediating embryo implantation, immune tolerance, nutritional supply and gas exchange. Human trophoblast stem cells (hTSCs) provide a platform to study lineage specification of placental tissues; however, the regulatory network controlling self-renewal remains elusive. Here, we present a single-cell atlas of human trophoblast development from zygote to mid-gestation together with single-cell profiling of hTSCs. We determine the transcriptional networks of trophoblast lineages in vivo and leverage probabilistic modelling to identify a role for MAPK signalling in trophoblast differentiation. Placenta- and blastoid-derived hTSCs consistently map between late trophectoderm and early cytotrophoblast, in contrast to blastoid-trophoblast, which correspond to trophectoderm. We functionally assess the requirement of the predicted cytotrophoblast network in an siRNA-screen and reveal 15 essential regulators for hTSC self-renewal, including MAZ, NFE2L3, TFAP2C, NR2F2 and CTNNB1. Our human trophoblast atlas provides a powerful analytical resource to delineate trophoblast cell fate acquisition, to elucidate transcription factors required for hTSC self-renewal and to gauge the developmental stage of in vitro cultured cells.
Collapse
Affiliation(s)
- Yutong Chen
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Site, Cambridge CB2 3EG, UK.,Centre for Trophoblast Research, University of Cambridge, Downing Site, Cambridge CB2 3EG, UK.,Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Dylan Siriwardena
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Site, Cambridge CB2 3EG, UK.,Centre for Trophoblast Research, University of Cambridge, Downing Site, Cambridge CB2 3EG, UK.,Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Christopher Penfold
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Site, Cambridge CB2 3EG, UK.,Centre for Trophoblast Research, University of Cambridge, Downing Site, Cambridge CB2 3EG, UK.,Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| | | | - Thorsten E Boroviak
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Site, Cambridge CB2 3EG, UK.,Centre for Trophoblast Research, University of Cambridge, Downing Site, Cambridge CB2 3EG, UK.,Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Puddicombe Way, Cambridge CB2 0AW, UK
| |
Collapse
|
20
|
Kobayashi N, Okae H, Hiura H, Kubota N, Kobayashi EH, Shibata S, Oike A, Hori T, Kikutake C, Hamada H, Kaji H, Suyama M, Bortolin-Cavaillé ML, Cavaillé J, Arima T. The microRNA cluster C19MC confers differentiation potential into trophoblast lineages upon human pluripotent stem cells. Nat Commun 2022; 13:3071. [PMID: 35654791 PMCID: PMC9163035 DOI: 10.1038/s41467-022-30775-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 05/13/2022] [Indexed: 02/08/2023] Open
Abstract
The first cell fate commitment during mammalian development is the specification of the inner cell mass and trophectoderm. This irreversible cell fate commitment should be epigenetically regulated, but the precise mechanism is largely unknown in humans. Here, we show that naïve human embryonic stem (hES) cells can transdifferentiate into trophoblast stem (hTS) cells, but primed hES cells cannot. Our transcriptome and methylome analyses reveal that a primate-specific miRNA cluster on chromosome 19 (C19MC) is active in naïve hES cells but epigenetically silenced in primed ones. Moreover, genome and epigenome editing using CRISPR/Cas systems demonstrate that C19MC is essential for hTS cell maintenance and C19MC-reactivated primed hES cells can give rise to hTS cells. Thus, we reveal that C19MC activation confers differentiation potential into trophoblast lineages on hES cells. Our findings are fundamental to understanding the epigenetic regulation of human early development and pluripotency. Little is known about the epigenetic mechanisms of the first cell fate commitment in humans. Here, the authors show that activation of the miRNA cluster C19MC confers differentiation potential into trophoblast lineages on human embryonic stem cells.
Collapse
Affiliation(s)
- Norio Kobayashi
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Hiroaki Okae
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan.
| | - Hitoshi Hiura
- Department of Bioscience, Faculty of Life Science, Tokyo University of Agriculture, Tokyo, 156-8502, Japan
| | - Naoto Kubota
- Division of Bioinformatics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Eri H Kobayashi
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Shun Shibata
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Akira Oike
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Takeshi Hori
- Department of Biomechanics, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, 101-0062, Japan
| | - Chie Kikutake
- Division of Bioinformatics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Hirotaka Hamada
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Hirokazu Kaji
- Department of Biomechanics, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo, 101-0062, Japan
| | - Mikita Suyama
- Division of Bioinformatics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Marie-Line Bortolin-Cavaillé
- Molecular, Cellular and Developmental biology department (MCD), Centre de Biologie Intégrative (CBI), University of Toulouse, CNRS, UPS, 31062, Toulouse, France
| | - Jérôme Cavaillé
- Molecular, Cellular and Developmental biology department (MCD), Centre de Biologie Intégrative (CBI), University of Toulouse, CNRS, UPS, 31062, Toulouse, France
| | - Takahiro Arima
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan.
| |
Collapse
|
21
|
Alexandrova M, Manchorova D, Dimova T. Immunity at maternal-fetal interface: KIR/HLA (Allo)recognition. Immunol Rev 2022; 308:55-76. [PMID: 35610960 DOI: 10.1111/imr.13087] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 04/28/2022] [Accepted: 05/09/2022] [Indexed: 12/15/2022]
Abstract
Both KIR and HLA are the most variable gene families in the human genome. The recognition of the semi-allogeneic embryo-derived trophoblasts by maternal decidual NK (dNK) cells is essential for the establishment of the functional placenta. This recognition is based on the KIR-HLA interactions and trophoblast expresses a specific HLA profile that constitutes classical polymorphic HLA-C and non-classical oligomorphic HLA-E, HLA-F, and HLA-G molecules. This review highlights some features of the KIR/HLA-C (allo)recognition by decidual NK (dNK) cells as a main immune cell population specifically enriched at maternal-fetal interface during human early pregnancy. How KIR/HLA-C axis operates in pregnancy disorders and in the context of transplacental infections is discussed as well. We summarized old and new data on dNK-cell functional plasticity, their selective expression of KIR and fetal maternal/paternal HLA-C haplotypes present. Results showed that KIR-HLA-C combinations and the corresponding axis operate differently in each pregnancy, determined by the variability of both maternal KIR haplotypes and fetus' maternal/paternal HLA-C allotype combinations. Moreover, the maturation of NK cells strongly depends on if or not HLA allotypes for certain KIR are present. We suggest that the unique KIR/HLA combinations reached in each pregnancy (normal and pathological) should be studied according to well-defined guidelines and unified methodologies to have comparable results ease to interpret and use in clinics.
Collapse
Affiliation(s)
- Marina Alexandrova
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Diana Manchorova
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Tanya Dimova
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, Sofia, Bulgaria
| |
Collapse
|
22
|
Generation of Cancer Stem/Initiating Cells by Cell-Cell Fusion. Int J Mol Sci 2022; 23:ijms23094514. [PMID: 35562905 PMCID: PMC9101717 DOI: 10.3390/ijms23094514] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/10/2022] [Accepted: 04/17/2022] [Indexed: 02/04/2023] Open
Abstract
CS/ICs have raised great expectations in cancer research and therapy, as eradication of this key cancer cell type is expected to lead to a complete cure. Unfortunately, the biology of CS/ICs is rather complex, since no common CS/IC marker has yet been identified. Certain surface markers or ALDH1 expression can be used for detection, but some studies indicated that cancer cells exhibit a certain plasticity, so CS/ICs can also arise from non-CS/ICs. Another problem is intratumoral heterogeneity, from which it can be inferred that different CS/IC subclones must be present in the tumor. Cell–cell fusion between cancer cells and normal cells, such as macrophages and stem cells, has been associated with the generation of tumor hybrids that can exhibit novel properties, such as an enhanced metastatic capacity and even CS/IC properties. Moreover, cell–cell fusion is a complex process in which parental chromosomes are mixed and randomly distributed among daughter cells, resulting in multiple, unique tumor hybrids. These, if they have CS/IC properties, may contribute to the heterogeneity of the CS/IC pool. In this review, we will discuss whether cell–cell fusion could also lead to the origin of different CS/ICs that may expand the overall CS/IC pool in a primary tumor.
Collapse
|
23
|
Yoshida K, Yano A, Kusama K, Ishikawa G, Tamura K. Alpha 1 Antitrypsin Regulates Trophoblast Syncytialization and Inflammatory Factor Expression. Int J Mol Sci 2022; 23:ijms23041955. [PMID: 35216073 PMCID: PMC8879717 DOI: 10.3390/ijms23041955] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/28/2022] [Accepted: 02/06/2022] [Indexed: 12/12/2022] Open
Abstract
The serine protease inhibitor alpha1-antitrypsin (A1AT) may possess protective functions of impaired organs in a manner independent of its protease inhibitor activity. A1AT expression has been shown to fluctuate in patients with pregnancy-induced hypertension, which suggests that A1AT may play a role in the syncytialization of villous trophoblasts. A1AT expression was knocked down in primary trophoblasts. RNA was extracted from these cells and subjected to RNA-sequencing analysis to determine the levels of expression of markers of syncytialization and inflammation. In addition, A1AT protein was localized in trophoblastic cells in placental tissues. Knockdown of A1AT upregulated the expression of FOSL1 and markers of syncytialization, as well as cell fusion, whereas overexpression of A1AT had the opposite effects. FOSL1 overexpression stimulated syncytialization, similar to the effects of A1AT knock down. Inhibitors of p38MAPK and JNK reduce the expression of inflammatory factors, whereas a p38MAPK inhibitor suppressed FOSL1 expression. Collectively, these findings indicated A1AT may negatively regulate inflammatory responses by controlling the activation of p38MAPK and JNK, and that p38MAPK mediates trophoblast syncytialization by altering FOSL1 expression. Therefore, a dysfunction in A1AT could be responsible for abnormal placental formation and pregnancy-associated disorders.
Collapse
Affiliation(s)
- Kanoko Yoshida
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (K.Y.); (A.Y.)
| | - Aruto Yano
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (K.Y.); (A.Y.)
| | - Kazuya Kusama
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (K.Y.); (A.Y.)
- Correspondence: (K.K.); (K.T.); Tel.: +81-42-676-4530 (K.K.); +81-42-676-4526 (K.T.)
| | - Gen Ishikawa
- Department of Obstetrics, Miyagi Children’s Hospital, Sendai 989-3126, Japan;
| | - Kazuhiro Tamura
- Department of Endocrine Pharmacology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (K.Y.); (A.Y.)
- Correspondence: (K.K.); (K.T.); Tel.: +81-42-676-4530 (K.K.); +81-42-676-4526 (K.T.)
| |
Collapse
|
24
|
Cao C, Dai Y, Wang Z, Zhao G, Duan H, Zhu X, Wang J, Zheng M, Weng Q, Wang L, Gou W, Zhang H, Li C, Liu D, Hu Y. The role of junctional adhesion molecule-C in trophoblast differentiation and function during normal pregnancy and preeclampsia. Placenta 2022; 118:55-65. [PMID: 35032792 DOI: 10.1016/j.placenta.2022.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/19/2021] [Accepted: 01/05/2022] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Junctional adhesion molecule-C (JAM-C) is an important regulator of many physiological processes, ranging from maintenance of tight junction integrity of epithelia to regulation of cell migration, homing and proliferation. Preeclampsia (PE) is a trophoblast-related syndrome with abnormal placentation and insufficient trophoblast invasion. However, the role of JAM-C in normal pregnancy and PE pathogenesis is unknown. METHODS The expression and location of JAM-C in placentas were determined by quantitative real-time PCR (qRT-PCR), western blot and immunohistochemistry. The expression of differentiation and invasion markers were detected by qRT-PCR or western blot. The effects of JAM-C on migration and invasion of trophoblasts were examined using wound-healing and invasion assays. Additionally, a mouse model was established by injection of JAM-C-positive adenovirus to explore the effects of JAM-C in vivo. RESULTS In normal pregnancy, JAM-C was preferentially expressed on cytotrophoblast (CTB) progenitors and progressively decreased when acquiring invasion properties with gestation advance. However, in PE patients, the expression of JAM-C was upregulated in extravillous trophoblasts (EVTs) and syncytiotrophoblasts (SynTs) of placentas. It was also demonstrated that JAM-C suppressed the differentiation of CTBs into EVTs in vitro. Consistently, JAM-C inhibited the migration and invasion capacities of EVTs through GSK3β/β-catenin signaling pathway. Importantly, Ad-JAMC-infected mouse model mimicked the phenotype of human PE. DISCUSSION JAM-C plays an important role in normal placentation and upregulated JAM-C in placentas contributes to PE development.
Collapse
Affiliation(s)
- Chenrui Cao
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yimin Dai
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhiyin Wang
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Guangfeng Zhao
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Honglei Duan
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Xiangyu Zhu
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Jingmei Wang
- Department of Pathology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Mingming Zheng
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Qiao Weng
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Limin Wang
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Wenjing Gou
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Haili Zhang
- Department of Obstetrics and Gynecology, The First People's Hospital of Mangya, Qinghai, China
| | - Chanjuan Li
- Department of Obstetrics, Women's Hospital of Nanjing Medical University, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Heath Care Hospital, Nanjing, China
| | - Dan Liu
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.
| | - Yali Hu
- Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
25
|
Kobayashi EH, Shibata S, Oike A, Kobayashi N, Hamada H, Okae H, Arima T. Genomic imprinting in human placentation. Reprod Med Biol 2022; 21:e12490. [PMID: 36465588 PMCID: PMC9713850 DOI: 10.1002/rmb2.12490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/25/2022] [Accepted: 11/10/2022] [Indexed: 12/02/2022] Open
Abstract
Background Genomic imprinting (GI) is a mammalian-specific epigenetic phenomenon that has been implicated in the evolution of the placenta in mammals. Methods Embryo transfer procedures and trophoblast stem (TS) cells were used to re-examine mouse placenta-specific GI genes. For the analysis of human GI genes, cytotrophoblast cells isolated from human placental tissues were used. Using human TS cells, the biological roles of human GI genes were examined. Main findings (1) Many previously identified mouse GI genes were likely to be falsely identified due to contaminating maternal cells. (2) Human placenta-specific GI genes were comprehensively determined, highlighting incomplete erasure of germline DNA methylation in the human placenta. (3) Human TS cells retained normal GI patterns. (4) Complete hydatidiform mole-derived TS cells were characterized by aberrant GI and enhanced trophoblastic proliferation. The maternally expressed imprinted gene p57KIP2 may be responsible for the enhanced proliferation. (5) The primate-specific microRNA cluster on chromosome 19, which is a placenta-specific GI gene, is essential for self-renewal and differentiation of human TS cells. Conclusion Genomic imprinting plays diverse and important roles in human placentation. Experimental analyses using TS cells suggest that the GI maintenance is necessary for normal placental development in humans.
Collapse
Affiliation(s)
- Eri H. Kobayashi
- Department of Informative GeneticsTohoku University School of MedicineSendaiJapan
| | - Shun Shibata
- Department of Informative GeneticsTohoku University School of MedicineSendaiJapan
| | - Akira Oike
- Department of Informative GeneticsTohoku University School of MedicineSendaiJapan
| | - Norio Kobayashi
- Department of Informative GeneticsTohoku University School of MedicineSendaiJapan
| | - Hirotaka Hamada
- Department of Informative GeneticsTohoku University School of MedicineSendaiJapan
| | - Hiroaki Okae
- Department of Informative GeneticsTohoku University School of MedicineSendaiJapan
| | - Takahiro Arima
- Department of Informative GeneticsTohoku University School of MedicineSendaiJapan
| |
Collapse
|
26
|
Kulus M, Sibiak R, Stefańska K, Zdun M, Wieczorkiewicz M, Piotrowska-Kempisty H, Jaśkowski JM, Bukowska D, Ratajczak K, Zabel M, Mozdziak P, Kempisty B. Mesenchymal Stem/Stromal Cells Derived from Human and Animal Perinatal Tissues-Origins, Characteristics, Signaling Pathways, and Clinical Trials. Cells 2021; 10:cells10123278. [PMID: 34943786 PMCID: PMC8699543 DOI: 10.3390/cells10123278] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/13/2021] [Accepted: 11/19/2021] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) are currently one of the most extensively researched fields due to their promising opportunity for use in regenerative medicine. There are many sources of MSCs, of which cells of perinatal origin appear to be an invaluable pool. Compared to embryonic stem cells, they are devoid of ethical conflicts because they are derived from tissues surrounding the fetus and can be safely recovered from medical waste after delivery. Additionally, perinatal MSCs exhibit better self-renewal and differentiation properties than those derived from adult tissues. It is important to consider the anatomy of perinatal tissues and the general description of MSCs, including their isolation, differentiation, and characterization of different types of perinatal MSCs from both animals and humans (placenta, umbilical cord, amniotic fluid). Ultimately, signaling pathways are essential to consider regarding the clinical applications of MSCs. It is important to consider the origin of these cells, referring to the anatomical structure of the organs of origin, when describing the general and specific characteristics of the different types of MSCs as well as the pathways involved in differentiation.
Collapse
Affiliation(s)
- Magdalena Kulus
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.K.); (K.R.)
| | - Rafał Sibiak
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (R.S.); (K.S.)
- Division of Reproduction, Department of Obstetrics, Gynecology, and Gynecologic Oncology, Poznan University of Medical Sciences, 60-535 Poznan, Poland
| | - Katarzyna Stefańska
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (R.S.); (K.S.)
| | - Maciej Zdun
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.Z.); (M.W.); (H.P.-K.)
| | - Maria Wieczorkiewicz
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.Z.); (M.W.); (H.P.-K.)
| | - Hanna Piotrowska-Kempisty
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.Z.); (M.W.); (H.P.-K.)
- Department of Toxicology, Poznan University of Medical Sciences, 60-631 Poznan, Poland
| | - Jędrzej M. Jaśkowski
- Department of Diagnostics and Clinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (J.M.J.); (D.B.)
| | - Dorota Bukowska
- Department of Diagnostics and Clinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (J.M.J.); (D.B.)
| | - Kornel Ratajczak
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.K.); (K.R.)
| | - Maciej Zabel
- Division of Anatomy and Histology, University of Zielona Gora, 65-046 Zielona Gora, Poland;
| | - Paul Mozdziak
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27695, USA;
| | - Bartosz Kempisty
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland; (M.K.); (K.R.)
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (R.S.); (K.S.)
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27695, USA;
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznan, Poland
- Correspondence:
| |
Collapse
|
27
|
Manzan-Martins C, Paulesu L. Impact of bisphenol A (BPA) on cells and tissues at the human materno-fetal interface. Tissue Cell 2021; 73:101662. [PMID: 34628212 DOI: 10.1016/j.tice.2021.101662] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 02/07/2023]
Abstract
Bisphenol A (BPA) is an endocrine disruptor extensively used in the production of polycarbonate plastics and epoxy resins and a component of liquid and food containers. It is a hazard in the prenatal period because of its presence in the placenta, fetal membranes, amniotic fluid, maternal and fetal blood and its ability to cross the placenta and reach the fetus. Estimation of the risk of BPA exposure during in utero life is extremely important in order to prevent complications of pregnancy and fetal growth. This review describes in vitro models of the human materno-fetal interface. It also outlines the effects of BPA at doses indicated as "physiological", namely at the concentrations found in the general population, and at "supraphysiological" and "subphysiological" doses, i.e. above and below the physiological range. This work will help clarify the discrepancies observed in studies on the effects of BPA on human reproduction and pregnancy, and it will be useful for the choice of appropriate in vitro models for future studies aimed at identifying the potential impact of BPA on specific functional processes.
Collapse
Affiliation(s)
| | - L Paulesu
- Department of Life Sciences, University of Siena, Siena, Italy.
| |
Collapse
|
28
|
Yart L, Roset Bahmanyar E, Cohen M, Martinez de Tejada B. Role of the Uteroplacental Renin-Angiotensin System in Placental Development and Function, and Its Implication in the Preeclampsia Pathogenesis. Biomedicines 2021; 9:biomedicines9101332. [PMID: 34680449 PMCID: PMC8533592 DOI: 10.3390/biomedicines9101332] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/13/2021] [Accepted: 09/21/2021] [Indexed: 11/23/2022] Open
Abstract
Placental development and function implicate important morphological and physiological adaptations to thereby ensure efficient maternal–fetal exchanges, as well as pregnancy-specific hormone secretion and immune modulation. Incorrect placental development can lead to severe pregnancy disorders, such as preeclampsia (PE), which endangers both the mother and the infant. The implication of the systemic renin–angiotensin system (RAS) in the pregnancy-related physiological changes is now well established. However, despite the fact that the local uteroplacental RAS has been described for several decades, its role in placental development and function seems to have been underestimated. In this review, we provide an overview of the multiple roles of the uteroplacental RAS in several cellular processes of placental development, its implication in the regulation of placental function during pregnancy, and the consequences of its dysregulation in PE pathogenesis.
Collapse
Affiliation(s)
- Lucile Yart
- Department of Pediatrics, Gynecology and Obstetrics, University Hospitals of Geneva, University of Geneva, 1211 Geneva, Switzerland; (L.Y.); (M.C.)
| | | | - Marie Cohen
- Department of Pediatrics, Gynecology and Obstetrics, University Hospitals of Geneva, University of Geneva, 1211 Geneva, Switzerland; (L.Y.); (M.C.)
| | - Begoña Martinez de Tejada
- Department of Pediatrics, Gynecology and Obstetrics, University Hospitals of Geneva, University of Geneva, 1211 Geneva, Switzerland; (L.Y.); (M.C.)
- Correspondence:
| |
Collapse
|
29
|
The Role and Clinical Interest of Extracellular Vesicles in Pregnancy and Ovarian Cancer. Biomedicines 2021; 9:biomedicines9091257. [PMID: 34572444 PMCID: PMC8464910 DOI: 10.3390/biomedicines9091257] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/14/2021] [Accepted: 09/14/2021] [Indexed: 12/21/2022] Open
Abstract
Ovarian cancer and pregnancy are two states in which the host immune system is exposed to novel antigens. Indeed, both the tumor and placenta must invade tissues, remodel vasculature to establish a robust blood supply, and evade detection by the immune system. Interestingly, tumor and placenta tissue use similar mechanisms to induce these necessary changes. One mediator is emerging as a key player in invasion, vascular remodeling, and immune evasion: extracellular vesicles (EVs). Many studies have identified EVs as a key mediator of cell-to-cell communication. Specifically, the cargo carried by EVs, which includes proteins, nucleic acids, and lipids, can interact with cells to induce changes in the target cell ranging from gene expression to migration and metabolism. EVs can promote cell division and tissue invasion, immunosuppression, and angiogenesis which are essential for both cancer and pregnancy. In this review, we examine the role of EVs in ovarian cancer metastasis, chemoresistance, and immune modulation. We then focus on the role of EVs in pregnancy with special attention on the vascular remodeling and regulation of the maternal immune system. Lastly, we discuss the clinical utility of EVs as markers and therapeutics for ovarian cancer and pre-eclampsia.
Collapse
|
30
|
Sun X, Tong X, Hao Y, Li C, Zhang Y, Pan Y, Dai Y, Liu L, Zhang T, Zhang S. Abnormal Cullin1 neddylation-mediated p21 accumulation participates in the pathogenesis of recurrent spontaneous abortion by regulating trophoblast cell proliferation and differentiation. Mol Hum Reprod 2021; 26:327-339. [PMID: 32186736 PMCID: PMC7227182 DOI: 10.1093/molehr/gaaa021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 02/29/2020] [Indexed: 01/07/2023] Open
Abstract
The study explores the role of neddylation in early trophoblast development and its alteration during the pathogenesis of recurrent spontaneous abortion (RSA). Immunofluorescence and western blot were conducted to evaluate the expression pattern of NEDD8 protein in the first-trimester placentas of healthy control and RSA patients. Neddylated-cullins, especially neddylated-cullin1, were downregulated and their substrate, p21, was accumulated in RSA samples. NEDD8 cytoplasmic recruitment was observed in extravillous trophoblast (EVT) progenitors of RSA placentas. Consistent with the results of clinical samples, neddylation inhibition using MLN4924 in trophoblast cell lines caused obvious p21 accumulation and free NEDD8 cytoplasmic recruitment. Further in vitro study demonstrated neddylation inhibition attenuated proliferation of Jeg-3 cells via p21 accumulation. Moreover, when trophoblast stem (TS) cells derived from first-trimester placentas were cultured for differentiation analyses. MLN4924 impaired the differentiation of TS cells towards EVTs by downregulating HLA-G and GATA3. p21 knockdown could partly rescue MLN4924-suppressed HLA-G and GATA3 expression. In conclusion, cullin1 neddylation-mediated p21 degradation is required for trophoblast proliferation and can affect trophoblast plasticity by affecting HLA-G and GATA3 expression. The results provide insights into the pathological mechanism of RSA and the biological regulation of trophoblast development.
Collapse
Affiliation(s)
- Xiaohe Sun
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Xiaomei Tong
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Yanqing Hao
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Chao Li
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Yinli Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Yibin Pan
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Yongdong Dai
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Liu Liu
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Tai Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Songying Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| |
Collapse
|
31
|
Díaz-Hernández I, Alecsandru D, García-Velasco JA, Domínguez F. Uterine natural killer cells: from foe to friend in reproduction. Hum Reprod Update 2021; 27:720-746. [PMID: 33528013 DOI: 10.1093/humupd/dmaa062] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/15/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Recurrent miscarriage and pre-eclampsia are common reproductive disorders, but their causes are often unknown. Recent evidence has provided new insight into immune system influences in reproductive disorders. A subset of lymphocytes of the innate immune system known as uterine natural killer (uNK) cells are now recognized as fundamental to achieving embryo implantation and successful pregnancy, but were initially attributed a bad reputation. Indeed, immune therapies have been developed to treat the 'exaggerated' immune response from uNK cells. These treatments have been based on studies of peripheral blood natural killer (pbNK) cells. However, uNK cells and pbNK cells have different phenotypic and functional characteristics. The functions of uNK cells are closely related to their interactions with the extravillous trophoblast cells (EVTs) and spiral arteries, which underlie an essential role in regulating vascular function, controlling trophoblast invasion and promoting placental development. EVTs express MHC molecules of class I HLA-C/E/G/F, while uNK cells express, among other receptors, killer cell immunoglobulin-like receptors (KIRs) that bind to HLA-C or CD94/NKG2A inhibitory receptors, and then bind HLA-E. Associations of certain KIR/HLA-C combinations with recurrent miscarriage, pre-eclampsia, and foetal growth restriction and the interactions between uNK cells, trophoblasts and vascular cells have led to the hypothesis that uNK cells may play a role in embryo implantation. OBJECTIVE AND RATIONALE Our objective was to review the evolution of our understanding of uNK cells, their functions, and their increasingly relevant role in reproduction. SEARCH METHODS Relevant literature through June 2020 was retrieved using Google Scholar and PubMed. Search terms comprised uNK cells, human pregnancy, reproductive failure, maternal KIR and HLA-C, HLA-E/G/F in EVT cells, angiogenic cytokines, CD56+ NK cells, spiral artery, oestrogen and progesterone receptors, KIR haplotype and paternal HLA-C2. OUTCOMES This review provides key insights into the evolving conceptualization of uNK cells, from their not-so-promising beginnings to now, when they are considered allies in reproduction. We synthesized current knowledge about uNK cells, their involvement in reproduction and their main functions in placental vascular remodeling and trophoblast invasion. One of the issues that this review presents is the enormous complexity involved in studying the immune system in reproduction. The complexity in the immunology of the maternal-foetal interface lies in the great variety of participating molecules, the processes and interactions that occur at different levels (molecular, cellular, tissue, etc.) and the great diversity of genetic combinations that are translated into different types of responses. WIDER IMPLICATIONS Insights into uNK cells could offer an important breakthrough for ART outcomes, since each patient could be assessed based on the combination of HLA and its receptors in their uNK cells, evaluating the critical interactions at the materno-foetal interface. However, owing to the technical challenges in studying uNK cells in vivo, there is still much knowledge to gain, particularly regarding their exact origin and functions. New studies using novel molecular and genetic approaches can facilitate the identification of mechanisms by which uNK cells interact with other cells at the materno-foetal interface, perhaps translating this knowledge into clinical applicability.
Collapse
Affiliation(s)
| | - Diana Alecsandru
- Department of Immunology and Department of Reproductive Endocrinology and Infertility, Instituto Valenciano de Infertilidad-Madrid, Rey Juan Carlos University (IVI), Madrid 28023, Spain
| | - Juan Antonio García-Velasco
- Department of Immunology and Department of Reproductive Endocrinology and Infertility, Instituto Valenciano de Infertilidad-Madrid, Rey Juan Carlos University (IVI), Madrid 28023, Spain
| | | |
Collapse
|
32
|
Molè MA, Coorens THH, Shahbazi MN, Weberling A, Weatherbee BAT, Gantner CW, Sancho-Serra C, Richardson L, Drinkwater A, Syed N, Engley S, Snell P, Christie L, Elder K, Campbell A, Fishel S, Behjati S, Vento-Tormo R, Zernicka-Goetz M. A single cell characterisation of human embryogenesis identifies pluripotency transitions and putative anterior hypoblast centre. Nat Commun 2021; 12:3679. [PMID: 34140473 PMCID: PMC8211662 DOI: 10.1038/s41467-021-23758-w] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 05/11/2021] [Indexed: 01/02/2023] Open
Abstract
Following implantation, the human embryo undergoes major morphogenetic transformations that establish the future body plan. While the molecular events underpinning this process are established in mice, they remain unknown in humans. Here we characterise key events of human embryo morphogenesis, in the period between implantation and gastrulation, using single-cell analyses and functional studies. First, the embryonic epiblast cells transition through different pluripotent states and act as a source of FGF signals that ensure proliferation of both embryonic and extra-embryonic tissues. In a subset of embryos, we identify a group of asymmetrically positioned extra-embryonic hypoblast cells expressing inhibitors of BMP, NODAL and WNT signalling pathways. We suggest that this group of cells can act as the anterior singalling centre to pattern the epiblast. These results provide insights into pluripotency state transitions, the role of FGF signalling and the specification of anterior-posterior axis during human embryo development.
Collapse
Affiliation(s)
- Matteo A Molè
- Department of Physiology, Development and Neuroscience, Mammalian Embryo and Stem Cell Group, University of Cambridge, Cambridge, UK
- Babraham Institute, Babraham Research Campus, Cambridge, UK
| | | | - Marta N Shahbazi
- Department of Physiology, Development and Neuroscience, Mammalian Embryo and Stem Cell Group, University of Cambridge, Cambridge, UK
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Antonia Weberling
- Department of Physiology, Development and Neuroscience, Mammalian Embryo and Stem Cell Group, University of Cambridge, Cambridge, UK
| | - Bailey A T Weatherbee
- Department of Physiology, Development and Neuroscience, Mammalian Embryo and Stem Cell Group, University of Cambridge, Cambridge, UK
| | - Carlos W Gantner
- Department of Physiology, Development and Neuroscience, Mammalian Embryo and Stem Cell Group, University of Cambridge, Cambridge, UK
| | | | - Lucy Richardson
- Herts & Essex Fertility Centre, Bishops College, Cheshunt, Herts, UK
| | - Abbie Drinkwater
- Herts & Essex Fertility Centre, Bishops College, Cheshunt, Herts, UK
| | - Najma Syed
- Herts & Essex Fertility Centre, Bishops College, Cheshunt, Herts, UK
| | - Stephanie Engley
- Herts & Essex Fertility Centre, Bishops College, Cheshunt, Herts, UK
| | | | | | | | | | - Simon Fishel
- CARE Fertility Group, Nottingham, UK
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Sam Behjati
- Wellcome Sanger Institute, Hinxton, UK.
- Cambridge University Hospital, NHS Foundation Trust, Cambridge, UK.
- Department of Paediatrics, University of Cambridge, Cambridge, UK.
| | | | - Magdalena Zernicka-Goetz
- Department of Physiology, Development and Neuroscience, Mammalian Embryo and Stem Cell Group, University of Cambridge, Cambridge, UK.
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
33
|
Adu-Gyamfi EA, Wang YX, Ding YB. The interplay between thyroid hormones and the placenta: a comprehensive review†. Biol Reprod 2021; 102:8-17. [PMID: 31494673 DOI: 10.1093/biolre/ioz182] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/21/2019] [Accepted: 09/05/2019] [Indexed: 12/13/2022] Open
Abstract
Thyroid hormones (THs) regulate a number of metabolic processes during pregnancy. After implantation, the placenta forms and enhances embryonic growth and development. Dysregulated maternal THs signaling has been observed in malplacentation-mediated pregnancy complications such as preeclampsia, miscarriage, and intrauterine growth restriction (IUGR), but the molecular mechanisms involved in this association have not been fully characterized. In this review, we have discussed THs signaling and its roles in trophoblast proliferation, trophoblast differentiation, trophoblast invasion of the decidua, and decidual angiogenesis. We have also explored the relationship between specific pregnancy complications and placental THs transporters, deiodinases, and THs receptors. In addition, we have examined the effects of specific endocrine disruptors on placental THs signaling. The available evidence indicates that THs signaling is involved in the formation and functioning of the placenta and serves as the basis for understanding the pathogenesis and pathophysiology of dysthyroidism-associated pregnancy complications such as preeclampsia, miscarriage, and IUGR.
Collapse
Affiliation(s)
- Enoch Appiah Adu-Gyamfi
- Department of Reproductive Sciences, School of Public Health, Chongqing Medical University, Chongqing, People's Republic of China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, People's Republic of China
| | - Ying-Xiong Wang
- Department of Reproductive Sciences, School of Public Health, Chongqing Medical University, Chongqing, People's Republic of China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, People's Republic of China
| | - Yu-Bin Ding
- Department of Reproductive Sciences, School of Public Health, Chongqing Medical University, Chongqing, People's Republic of China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
34
|
Extracellularly Released Calreticulin Induced by Endoplasmic Reticulum Stress Impairs Syncytialization of Cytotrophoblast Model BeWo Cells. Cells 2021; 10:cells10061305. [PMID: 34073978 PMCID: PMC8225044 DOI: 10.3390/cells10061305] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 12/12/2022] Open
Abstract
The pregnancy-specific syndrome preeclampsia is a major cause of maternal mortality throughout the world. The initial insult resulting in the development of preeclampsia is inadequate trophoblast invasion, which may lead to reduced maternal perfusion of the placenta and placental dysfunction, such as insufficient trophoblast syncytialization. Endoplasmic reticulum (ER) stress has been implicated in the pathology of preeclampsia and serves as the major risk factor. Our previous studies suggested critical roles of calreticulin (CRT), which is an ER-resident stress response protein, in extravillous trophoblast invasion and cytotrophoblast syncytialization. Here, we studied the mechanism by which ER stress exposes the placenta to the risk of preeclampsia. We found that CRT was upregulated in the serum samples, but not in the placental specimens, from preeclamptic women. By using BeWo cells, an established model of cytotrophoblasts that syncytialize in the presence of forskolin, we demonstrated that thapsigargin-induced ER stress caused extracellular release of CRT from BeWo cells and that the extracellular CRT suppressed forskolin-induced release of β-human chorionic gonadotropin and altered subcellular localization of E-cadherin, which is a key adhesion molecule associated with syncytialization. Our results together provide evidence that induction of ER stress leads to extracellular CRT release, which may contribute to placental dysfunction by suppressing cytotrophoblast syncytialization.
Collapse
|
35
|
Syncytiotrophoblast stress in early onset preeclampsia: The issues perpetuating the syndrome. Placenta 2021; 113:57-66. [PMID: 34053733 DOI: 10.1016/j.placenta.2021.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 04/26/2021] [Accepted: 05/14/2021] [Indexed: 01/08/2023]
Abstract
Preeclampsia is a pregnancy-specific syndrome characterized by a sudden increase in blood pressure accompanied by proteinuria and/or maternal multi-system damage associated to poor fetal outcome. In early-onset preeclampsia, utero-placental perfusion is altered, causing constant and progressive damage to the syncytiotrophoblast, generating syncytiotrophoblast stress. The latter leads to the detachment and release of syncytiotrophoblast fragments, anti-angiogenic factors and pro-inflammatory molecules into maternal circulation, resulting in the emergence and persistence of the characteristic symptoms of this syndrome during pregnancy. Therefore, understanding the origin and consequences of syncytiotrophoblast stress in preeclampsia is vital to develop new therapeutic alternatives, focused on reducing the burden of this syndrome. In this review, we describe five central characteristics of syncytial stress that should be targeted or prevented in order to reduce preeclampsia symptoms: histological alterations, syncytiotrophoblast damage, antiangiogenic protein export, placental deportation, and altered syncytiotrophoblast turnover. Therapeutic management of these characteristics may improve maternal and fetal outcomes.
Collapse
|
36
|
Two distinct trophectoderm lineage stem cells from human pluripotent stem cells. J Biol Chem 2021; 296:100386. [PMID: 33556374 PMCID: PMC7948510 DOI: 10.1016/j.jbc.2021.100386] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 01/27/2021] [Accepted: 02/03/2021] [Indexed: 01/08/2023] Open
Abstract
The trophectoderm layer of the blastocyst-stage embryo is the precursor for all trophoblast cells in the placenta. Human trophoblast stem (TS) cells have emerged as an attractive tool for studies on early trophoblast development. However, the use of TS cell models is constrained by the limited genetic diversity of existing TS cell lines and restrictions on using human fetal tissue or embryos needed to generate additional lines. Here we report the derivation of two distinct stem cell types of the trophectoderm lineage from human pluripotent stem cells. Analogous to villous cytotrophoblasts in vivo, the first is a CDX2- stem cell comparable with placenta-derived TS cells—they both exhibit identical expression of key markers, are maintained in culture and differentiate under similar conditions, and share high transcriptome similarity. The second is a CDX2+ stem cell with distinct cell culture requirements, and differences in gene expression and differentiation, relative to CDX2- stem cells. Derivation of TS cells from pluripotent stem cells will significantly enable construction of in vitro models for normal and pathological placental development.
Collapse
|
37
|
Pan Y, Yan L, Chen Q, Wei C, Dai Y, Tong X, Zhu H, Lu M, Zhang Y, Jin X, Zhang T, Lin X, Zhou F, Zhang S. Dysfunction of Shh signaling activates autophagy to inhibit trophoblast motility in recurrent miscarriage. Exp Mol Med 2021; 53:52-66. [PMID: 33390589 PMCID: PMC8080798 DOI: 10.1038/s12276-020-00530-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 09/23/2020] [Accepted: 10/20/2020] [Indexed: 01/29/2023] Open
Abstract
In early pregnancy, the placenta anchors the conceptus and supports embryonic development and survival. This study aimed to investigate the underlying functions of Shh signaling in recurrent miscarriage (RM), a serious disorder of pregnancy. In the present study, Shh and Gli2 were mainly observed in cytotrophoblasts (CTBs), Ptch was mainly observed in syncytiotrophoblasts (STBs), and Smo and Gli3 were expressed in both CTBs and STBs. Shh signaling was significantly impaired in human placenta tissue from recurrent miscarriage patients compared to that of gestational age-matched normal controls. VEGF-A and CD31 protein levels were also significantly decreased in recurrent miscarriage patients. Furthermore, inhibition of Shh signaling impaired the motility of JAR cells by regulating the expression of Gli2 and Gli3. Intriguingly, inhibition of Shh signaling also triggered autophagy and autolysosome accumulation. Additionally, knockdown of BECN1 reversed Gant61-induced motility inhibition. In conclusion, our results showed that dysfunction of Shh signaling activated autophagy to inhibit trophoblast motility, which suggests the Shh pathway and autophagy as potential targets for RM therapy.
Collapse
Affiliation(s)
- Yibin Pan
- grid.13402.340000 0004 1759 700XAssisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Lili Yan
- grid.13402.340000 0004 1759 700XAssisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China ,Beilun District Hospital of Traditional Chinese Medicine, Ningbo City, Zhejiang China
| | - Qiaoqiao Chen
- grid.13402.340000 0004 1759 700XAssisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Cheng Wei
- grid.13402.340000 0004 1759 700XAssisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Yongdong Dai
- grid.13402.340000 0004 1759 700XAssisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Xiaomei Tong
- grid.13402.340000 0004 1759 700XAssisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Haiyan Zhu
- grid.13402.340000 0004 1759 700XAssisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Meifei Lu
- grid.13402.340000 0004 1759 700XDepartment of Pharmacy, The Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanling Zhang
- grid.13402.340000 0004 1759 700XAssisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Xiaoying Jin
- grid.13402.340000 0004 1759 700XAssisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Tai Zhang
- grid.13402.340000 0004 1759 700XAssisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Xiaona Lin
- grid.13402.340000 0004 1759 700XAssisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Feng Zhou
- grid.13402.340000 0004 1759 700XAssisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Songying Zhang
- grid.13402.340000 0004 1759 700XAssisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China ,Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| |
Collapse
|
38
|
Lok C, Frijstein M, van Trommel N. Clinical presentation and diagnosis of Gestational Trophoblastic Disease. Best Pract Res Clin Obstet Gynaecol 2020; 74:42-52. [PMID: 33422446 DOI: 10.1016/j.bpobgyn.2020.12.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/05/2020] [Accepted: 12/01/2020] [Indexed: 12/14/2022]
Abstract
Gestational trophoblastic disease (GTD) is a heterogeneous group of pregnancy-related disorders characterized by abnormal proliferation of trophoblastic tissue. It encompasses the premalignant partial and complete hydatidiform mole but also the malignant invasive mole, choriocarcinoma, placental-site trophoblastic tumor, and epithelioid trophoblastic tumor. The clinical presentation changed to earlier detection after the introduction of first trimester ultrasounds. Patients are often asymptomatic, but vaginal bleeding continues to be the most common presenting symptom. Other symptoms can develop in the case of metastatic disease. Ultrasound, serum human chorionic gonadotrophin, and sometimes additional imaging such as CT, MRI, or PET can confirm the diagnosis and stage of disease. Familiarity with the pathogenesis, classification, imaging features, and treatment of GTD facilitates diagnosis and appropriate management.
Collapse
Affiliation(s)
- Christianne Lok
- Department of Gynaecologic Oncology, Center of Gynecologic Oncology Amsterdam, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands.
| | - Minke Frijstein
- Department of Gynaecologic Oncology, Center of Gynecologic Oncology Amsterdam, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands.
| | - Nienke van Trommel
- Department of Gynaecologic Oncology, Center of Gynecologic Oncology Amsterdam, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands.
| |
Collapse
|
39
|
Grisaru-Granovsky S, Kumar Nag J, Zakar L, Rudina T, Lal Gupta C, Maoz M, Kozlova D, Bar-Shavit R. PAR 1&2 driven placenta EVT invasion act via LRP5/6 as coreceptors. FASEB J 2020; 34:15701-15717. [PMID: 33136328 DOI: 10.1096/fj.202000306r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 06/29/2020] [Accepted: 07/06/2020] [Indexed: 12/17/2022]
Abstract
While the involvement of protease-activated receptors (PARs) in the physiological regulation of human placenta development, as in tumor biology, is recognized, the molecular pathway is unknown. We evaluated the impact of PAR1 and PAR2 function in cytotrophoblast (CTB) proliferation and invasion in a system of extravillous trophoblast (EVT) organ culture and in human cell-lines. Activation of PAR1 - and PAR2 -induced EVT invasion and proliferation, while the shRNA silencing of low-density lipoprotein receptor-related protein 5/6 (LRP5/6) inhibited these processes. PAR1 and PAR2 effectively induce β-catenin stabilization in a manner similar to that shown for the canonical β-catenin stabilization pathway yet independent of Wnts. Immunoprecipitation analyses and protein-protein docking demonstrated the co-association between either PAR1 or PAR2 with LRP5/6 forming an axis of PAR-LRP5/6-Axin. Noticeably, in PAR1 -PAR2 heterodimers a dominant role is assigned to PAR2 over PAR1 as shown by inhibition of PAR1 -induced β-catenin levels, and Dvl nuclear localization. This inhibition takes place either by shRNA silenced hPar2 or in the presence of a TrPAR2 devoid its cytoplasmic tail. Indeed, TrPAR2 cannot form the PAR1 -PAR2 complex, obstructing thereby the flow of signals downstream. Elucidation of the mechanism of PAR-induced invasion contributes to therapeutic options highlighting key partners in the process.
Collapse
Affiliation(s)
- Sorina Grisaru-Granovsky
- Department of Obstetrics and Gynecology, Hebrew-University, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Jeetendra Kumar Nag
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Liat Zakar
- Department of Obstetrics and Gynecology, Hebrew-University, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Tatyana Rudina
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Chhedi Lal Gupta
- Institute of Soil, Water and Environmental Sciences, Volcani Research Center, Agriculture Research Organization, Rishon Lezion, Israel
| | - Myriam Maoz
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Daria Kozlova
- Department of Obstetrics and Gynecology, Hebrew-University, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Rachel Bar-Shavit
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
40
|
Liu H, Wang L, Wang Y, Zhu Q, Aldo P, Ding J, Mor G, Liao A. Establishment and characterization of a new human first trimester Trophoblast cell line, AL07. Placenta 2020; 100:122-132. [PMID: 32927240 PMCID: PMC8237240 DOI: 10.1016/j.placenta.2020.08.013] [Citation(s) in RCA: 8] [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: 02/26/2020] [Revised: 07/20/2020] [Accepted: 08/12/2020] [Indexed: 11/21/2022]
Abstract
INTRODUCTION The limited cell number of primary trophoblasts and contamination of trophoblast cell lines promote us to develop a novel stable trophoblast cell line. METHOD OF STUDY Primary trophoblast cells were isolated from first-trimester placenta and telomerase-induced immortalization was used to immortalize these cells. Subsets of cells were then evaluated by flow cytometry using CK7, HLA-G, CD45 and CD14, specific markers for trophoblast cells, extra-villous trophoblast, pan leucocyte and monocyte/macrophage, respectively. Immunofluorescence staining and immunocytochemistry were used to detect CK7 expression in trophoblast cells. The level of secreted human Chorionic Gonadotropin (hCG) was measured by electrochemiluminescence (ECL). The Bio-Plex MAGPIX System was used to analyze the cytokines and chemokines produced by AL07 cell line. RESULTS We were able to isolate primary trophoblast cells from several first-trimester placentas. One clone, AL07 trophoblast cells, isolated from a week 7 placenta, was morphologically stable and positive for the expression of CK7 by immunofluorescence and immunocytochemistry staining. Characterization of AL07 cells reveled that they are CD45 or CD14 negative and had constitutive secretion of hCG and low HLA-G expression. Furthermore, clone AL07 secret high levels of several cytokines and chemokines, including IL-6, IL-8 and VEGF, and moderately secreted MCP-1 IP-10 and RANTES. DISCUSSION We report the successful isolation, immortalization and characterization of AL07 cells, a novel cell clone isolated from first trimester human placenta. The clone is free of contamination of immune cells, and exhibits similar cytokine profile as other trophoblast cell lines. This new cytotrophoblast-like AL07 cell, can be a valuable tool for in-vitro trophoblast studies in the future.
Collapse
Affiliation(s)
- Hong Liu
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Liling Wang
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Yan Wang
- Department of Obstetrics and Gynecology, Maternal and Child Health Hospital of Hubei Province, Wuhan, China
| | - Qian Zhu
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Paulomi Aldo
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Jiahui Ding
- C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, USA
| | - Gil Mor
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China; C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Aihua Liao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| |
Collapse
|
41
|
Horii M, Bui T, Touma O, Cho HY, Parast MM. An Improved Two-Step Protocol for Trophoblast Differentiation of Human Pluripotent Stem Cells. ACTA ACUST UNITED AC 2020; 50:e96. [PMID: 31479595 DOI: 10.1002/cpsc.96] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We previously established a two-step protocol for differentiation of human pluripotent stem cells (hPSCs) into trophoblasts, using a StemPro-based minimal medium (EMIM) with bone morphogenetic protein-4 (BMP4). This protocol was suboptimal, resulting in induction of mixed mesoderm and trophoblast markers. Furthermore, adapting hPSCs to StemPro has proven difficult, and prolonged culture in this medium has been shown to promote genomic instability. Therefore, we moved on to the use of new media, including E8, and most recently, StemFlex, for rapid adaptation from feeder to non-feeder conditions. In the new protocol, we have incorporated the WNT inhibitor IWP2 into the first step, resulting in uniform differentiation of hPSCs into cytotrophoblast (CTB)-like cells, without induction of the mesoderm lineage. We also show that, at the end of the second step, there are distinct populations of terminally differentiated multinucleated human chorionic gonadotropin (hCG)-producing syncytiotrophoblast (STB) and HLAG+ extravillous trophoblast (EVT)-like cells. © 2019 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Mariko Horii
- Department of Pathology, University of California San Diego, La Jolla, California.,Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Tony Bui
- Department of Pathology, University of California San Diego, La Jolla, California.,Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Ojeni Touma
- Department of Pathology, University of California San Diego, La Jolla, California.,Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Hee Young Cho
- Department of Pathology, University of California San Diego, La Jolla, California.,Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Mana M Parast
- Department of Pathology, University of California San Diego, La Jolla, California.,Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| |
Collapse
|
42
|
Horii M, Touma O, Bui T, Parast MM. Modeling human trophoblast, the placental epithelium at the maternal fetal interface. Reproduction 2020; 160:R1-R11. [PMID: 32485667 PMCID: PMC7286067 DOI: 10.1530/rep-19-0428] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 04/17/2020] [Indexed: 12/14/2022]
Abstract
Appropriate human trophoblast lineage specification and differentiation is crucial for the establishment of normal placentation and maintenance of pregnancy. However, due to the lack of proper modeling systems, the molecular mechanisms of these processes are still largely unknown. Much of the early studies in this area have been based on animal models and tumor-derived trophoblast cell lines, both of which are suboptimal for modeling this unique human organ. Recent advances in regenerative and stem cell biology methods have led to development of novel in vitro model systems for studying human trophoblast. These include derivation of human embryonic and induced pluripotent stem cells and establishment of methods for the differentiation of these cells into trophoblast, as well as the more recent derivation of human trophoblast stem cells. In addition, advances in culture conditions, from traditional two-dimensional monolayer culture to 3D culturing systems, have led to development of trophoblast organoid and placenta-on-a-chip model, enabling us to study human trophoblast function in context of more physiologically accurate environment. In this review, we will discuss these various model systems, with a focus on human trophoblast, and their ability to help elucidate the key mechanisms underlying placental development and function. This review focuses on model systems of human trophoblast differentiation, including advantages and limitations of stem cell-based culture, trophoblast organoid, and organ-on-a-chip methods and their applications in understanding placental development and disease.
Collapse
Affiliation(s)
- Mariko Horii
- Department of Pathology, University of California San Diego, La Jolla, California, USA
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California, USA
| | - Ojeni Touma
- Department of Pathology, University of California San Diego, La Jolla, California, USA
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California, USA
| | - Tony Bui
- Department of Pathology, University of California San Diego, La Jolla, California, USA
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California, USA
| | - Mana M Parast
- Department of Pathology, University of California San Diego, La Jolla, California, USA
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
43
|
H19 regulates angiogenic capacity of extravillous trophoblasts by H19/miR-106a-5p/VEGFA axis. Arch Gynecol Obstet 2020; 301:671-679. [DOI: 10.1007/s00404-020-05469-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 02/13/2020] [Indexed: 02/07/2023]
|
44
|
Dong C, Beltcheva M, Gontarz P, Zhang B, Popli P, Fischer LA, Khan SA, Park KM, Yoon EJ, Xing X, Kommagani R, Wang T, Solnica-Krezel L, Theunissen TW. Derivation of trophoblast stem cells from naïve human pluripotent stem cells. eLife 2020; 9:e52504. [PMID: 32048992 PMCID: PMC7062471 DOI: 10.7554/elife.52504] [Citation(s) in RCA: 184] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 02/11/2020] [Indexed: 12/24/2022] Open
Abstract
Naïve human pluripotent stem cells (hPSCs) provide a unique experimental platform of cell fate decisions during pre-implantation development, but their lineage potential remains incompletely characterized. As naïve hPSCs share transcriptional and epigenomic signatures with trophoblast cells, it has been proposed that the naïve state may have enhanced predisposition for differentiation along this extraembryonic lineage. Here we examined the trophoblast potential of isogenic naïve and primed hPSCs. We found that naïve hPSCs can directly give rise to human trophoblast stem cells (hTSCs) and undergo further differentiation into both extravillous and syncytiotrophoblast. In contrast, primed hPSCs do not support hTSC derivation, but give rise to non-self-renewing cytotrophoblasts in response to BMP4. Global transcriptome and chromatin accessibility analyses indicate that hTSCs derived from naïve hPSCs are similar to blastocyst-derived hTSCs and acquire features of post-implantation trophectoderm. The derivation of hTSCs from naïve hPSCs will enable elucidation of early mechanisms that govern normal human trophoblast development and associated pathologies.
Collapse
Affiliation(s)
- Chen Dong
- Department of Developmental Biology, Washington University School of MedicineSt. LouisUnited States
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
| | - Mariana Beltcheva
- Department of Developmental Biology, Washington University School of MedicineSt. LouisUnited States
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
| | - Paul Gontarz
- Department of Developmental Biology, Washington University School of MedicineSt. LouisUnited States
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
| | - Bo Zhang
- Department of Developmental Biology, Washington University School of MedicineSt. LouisUnited States
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
| | - Pooja Popli
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of MedicineSt. LouisUnited States
| | - Laura A Fischer
- Department of Developmental Biology, Washington University School of MedicineSt. LouisUnited States
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
| | - Shafqat A Khan
- Department of Developmental Biology, Washington University School of MedicineSt. LouisUnited States
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
| | - Kyoung-mi Park
- Department of Developmental Biology, Washington University School of MedicineSt. LouisUnited States
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
| | - Eun-Ja Yoon
- Department of Developmental Biology, Washington University School of MedicineSt. LouisUnited States
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
| | - Xiaoyun Xing
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
- Department of Genetics, Center for Genome Sciences & Systems Biology, Washington University School of MedicineSt. LouisUnited States
| | - Ramakrishna Kommagani
- Department of Obstetrics and Gynecology, Center for Reproductive Health Sciences, Washington University School of MedicineSt. LouisUnited States
| | - Ting Wang
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
- Department of Genetics, Center for Genome Sciences & Systems Biology, Washington University School of MedicineSt. LouisUnited States
| | - Lilianna Solnica-Krezel
- Department of Developmental Biology, Washington University School of MedicineSt. LouisUnited States
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
| | - Thorold W Theunissen
- Department of Developmental Biology, Washington University School of MedicineSt. LouisUnited States
- Center of Regenerative Medicine, Washington University School of MedicineSt. LouisUnited States
| |
Collapse
|
45
|
Szilagyi A, Gelencser Z, Romero R, Xu Y, Kiraly P, Demeter A, Palhalmi J, Gyorffy BA, Juhasz K, Hupuczi P, Kekesi KA, Meinhardt G, Papp Z, Draghici S, Erez O, Tarca AL, Knöfler M, Than NG. Placenta-Specific Genes, Their Regulation During Villous Trophoblast Differentiation and Dysregulation in Preterm Preeclampsia. Int J Mol Sci 2020; 21:ijms21020628. [PMID: 31963593 PMCID: PMC7013556 DOI: 10.3390/ijms21020628] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 12/15/2022] Open
Abstract
The human placenta maintains pregnancy and supports the developing fetus by providing nutrition, gas-waste exchange, hormonal regulation, and an immunological barrier from the maternal immune system. The villous syncytiotrophoblast carries most of these functions and provides the interface between the maternal and fetal circulatory systems. The syncytiotrophoblast is generated by the biochemical and morphological differentiation of underlying cytotrophoblast progenitor cells. The dysfunction of the villous trophoblast development is implicated in placenta-mediated pregnancy complications. Herein, we describe gene modules and clusters involved in the dynamic differentiation of villous cytotrophoblasts into the syncytiotrophoblast. During this process, the immune defense functions are first established, followed by structural and metabolic changes, and then by peptide hormone synthesis. We describe key transcription regulatory molecules that regulate gene modules involved in placental functions. Based on transcriptomic evidence, we infer how villous trophoblast differentiation and functions are dysregulated in preterm preeclampsia, a life-threatening placenta-mediated obstetrical syndrome for the mother and fetus. In the conclusion, we uncover the blueprint for villous trophoblast development and its impairment in preterm preeclampsia, which may aid in the future development of non-invasive biomarkers for placental functions and early identification of women at risk for preterm preeclampsia as well as other placenta-mediated pregnancy complications.
Collapse
Affiliation(s)
- Andras Szilagyi
- Systems Biology of Reproduction Lendulet Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (A.S.); (Z.G.); (P.K.); (A.D.); (J.P.); (K.J.)
| | - Zsolt Gelencser
- Systems Biology of Reproduction Lendulet Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (A.S.); (Z.G.); (P.K.); (A.D.); (J.P.); (K.J.)
| | - Roberto Romero
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD 20692, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (O.E.); (A.L.T.)
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
- Detroit Medical Center, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Florida International University, Miami, FL 33199, USA
| | - Yi Xu
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD 20692, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (O.E.); (A.L.T.)
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Peter Kiraly
- Systems Biology of Reproduction Lendulet Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (A.S.); (Z.G.); (P.K.); (A.D.); (J.P.); (K.J.)
| | - Amanda Demeter
- Systems Biology of Reproduction Lendulet Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (A.S.); (Z.G.); (P.K.); (A.D.); (J.P.); (K.J.)
| | - Janos Palhalmi
- Systems Biology of Reproduction Lendulet Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (A.S.); (Z.G.); (P.K.); (A.D.); (J.P.); (K.J.)
| | - Balazs A. Gyorffy
- Laboratory of Proteomics, Institute of Biology, Eotvos Lorand University, H-1117 Budapest, Hungary; (B.A.G.); (K.A.K.)
| | - Kata Juhasz
- Systems Biology of Reproduction Lendulet Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (A.S.); (Z.G.); (P.K.); (A.D.); (J.P.); (K.J.)
| | - Petronella Hupuczi
- Maternity Private Clinic of Obstetrics and Gynecology, H-1126 Budapest, Hungary; (P.H.); (Z.P.)
| | - Katalin Adrienna Kekesi
- Laboratory of Proteomics, Institute of Biology, Eotvos Lorand University, H-1117 Budapest, Hungary; (B.A.G.); (K.A.K.)
- Department of Physiology and Neurobiology, Eotvos Lorand University, H-1117 Budapest, Hungary
| | - Gudrun Meinhardt
- Department of Obstetrics and Gynecology, Reproductive Biology Unit, Medical University of Vienna, Vienna A-1090, Austria; (G.M.); (M.K.)
| | - Zoltan Papp
- Maternity Private Clinic of Obstetrics and Gynecology, H-1126 Budapest, Hungary; (P.H.); (Z.P.)
- Department of Obstetrics and Gynecology, Semmelweis University, H-1088 Budapest, Hungary
| | - Sorin Draghici
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI 48202, USA;
| | - Offer Erez
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD 20692, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (O.E.); (A.L.T.)
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel
| | - Adi Laurentiu Tarca
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD 20692, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (O.E.); (A.L.T.)
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Martin Knöfler
- Department of Obstetrics and Gynecology, Reproductive Biology Unit, Medical University of Vienna, Vienna A-1090, Austria; (G.M.); (M.K.)
| | - Nandor Gabor Than
- Systems Biology of Reproduction Lendulet Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (A.S.); (Z.G.); (P.K.); (A.D.); (J.P.); (K.J.)
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, MD 20692, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (O.E.); (A.L.T.)
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Maternity Private Clinic of Obstetrics and Gynecology, H-1126 Budapest, Hungary; (P.H.); (Z.P.)
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary
- Correspondence: ; Tel.: +36-1-382-6788
| |
Collapse
|
46
|
Loss of p57 KIP2 expression confers resistance to contact inhibition in human androgenetic trophoblast stem cells. Proc Natl Acad Sci U S A 2019; 116:26606-26613. [PMID: 31792181 PMCID: PMC6936680 DOI: 10.1073/pnas.1916019116] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Complete hydatidiform moles (CHMs) develop from androgenetic conceptuses and are characterized by enhanced proliferation of trophoblast cells and a significantly higher risk of trophoblast tumors. Loss of the maternal genome and duplication of the paternal genome are considered to be responsible for the phenotype, but the detailed mechanism remains unclear. Here, we report the derivation of trophoblast stem (TS) cells from CHMs. These cells have reduced sensitivity to contact inhibition of cell proliferation and exhibit aberrant expression of imprinted genes, which are expressed from only 1 parental allele. We also reveal that the maternally expressed imprinted gene p57KIP2 would be responsible for the enhanced proliferation of CHM-derived TS cells. Our findings provide an insight into the pathogenesis of CHMs. A complete hydatidiform mole (CHM) is androgenetic in origin and characterized by enhanced trophoblastic proliferation and the absence of fetal tissue. In 15 to 20% of cases, CHMs are followed by malignant gestational trophoblastic neoplasms including choriocarcinoma. Aberrant genomic imprinting may be responsible for trophoblast hypertrophy in CHMs, but the detailed mechanisms are still elusive, partly due to the lack of suitable animal or in vitro models. We recently developed a culture system of human trophoblast stem (TS) cells. In this study, we apply this system to CHMs for a better understanding of their molecular pathology. CHM-derived TS cells, designated as TSmole cells, are morphologically similar to biparental TS (TSbip) cells and express TS-specific markers such as GATA3, KRT7, and TFAP2C. Interestingly, TSmole cells have a growth advantage over TSbip cells only after they reach confluence. We found that p57KIP2, a maternally expressed gene encoding a cyclin-dependent kinase inhibitor, is strongly induced by increased cell density in TSbip cells, but not in TSmole cells. Knockout and overexpression studies suggest that loss of p57KIP2 expression would be the major cause of the reduced sensitivity to contact inhibition in CHMs. Our findings shed light on the molecular mechanism underlying the pathogenesis of CHMs and could have broad implications in tumorigenesis beyond CHMs because silencing of p57KIP2 is frequently observed in a variety of human tumors.
Collapse
|
47
|
Lv B, An Q, Zeng Q, Zhang X, Lu P, Wang Y, Zhu X, Ji Y, Fan G, Xue Z. Single-cell RNA sequencing reveals regulatory mechanism for trophoblast cell-fate divergence in human peri-implantation conceptuses. PLoS Biol 2019; 17:e3000187. [PMID: 31596842 PMCID: PMC6802852 DOI: 10.1371/journal.pbio.3000187] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 10/21/2019] [Accepted: 09/12/2019] [Indexed: 12/15/2022] Open
Abstract
Multipotent trophoblasts undergo dynamic morphological movement and cellular differentiation after conceptus implantation to generate placenta. However, the mechanism controlling trophoblast development and differentiation during peri-implantation development in human remains elusive. In this study, we modeled human conceptus peri-implantation development from blastocyst to early postimplantation stages by using an in vitro coculture system and profiled the transcriptome of 476 individual trophoblast cells from these conceptuses. We revealed the genetic networks regulating peri-implantation trophoblast development. While determining when trophoblast differentiation happens, our bioinformatic analysis identified T-box transcription factor 3 (TBX3) as a key regulator for the differentiation of cytotrophoblast (CT) into syncytiotrophoblast (ST). The function of TBX3 in trophoblast differentiation is then validated by a loss-of-function experiment. In conclusion, our results provided a valuable resource to study the regulation of trophoblasts development and differentiation during human peri-implantation development.
Collapse
Affiliation(s)
- Bo Lv
- Department of Regenerative Medicine, School of Medicine, Tongji University, Shanghai, China
| | - Qin An
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, State of California, United States of America
| | - Qiao Zeng
- Center of Reproductive Medicine of Ji’an Maternal and Child Health Hospital, Ji’an, Jiangxi, China
| | - Xunyi Zhang
- Reproductive Medicine Center, Tongji Hospital, Tongji University, Shanghai, China
| | - Ping Lu
- Department of Regenerative Medicine, School of Medicine, Tongji University, Shanghai, China
| | - Yanqiu Wang
- Reproductive Medicine Center, Tongji Hospital, Tongji University, Shanghai, China
| | - Xianmin Zhu
- Shanghai Pulmonary Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yazhong Ji
- Reproductive Medicine Center, Tongji Hospital, Tongji University, Shanghai, China
- * E-mail: (ZX); (GF); (YJ)
| | - Guoping Fan
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, State of California, United States of America
- * E-mail: (ZX); (GF); (YJ)
| | - Zhigang Xue
- Department of Regenerative Medicine, School of Medicine, Tongji University, Shanghai, China
- Reproductive Medicine Center, Tongji Hospital, Tongji University, Shanghai, China
- * E-mail: (ZX); (GF); (YJ)
| |
Collapse
|
48
|
Placental structure in gestational diabetes mellitus. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165535. [PMID: 31442531 DOI: 10.1016/j.bbadis.2019.165535] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/11/2019] [Accepted: 08/15/2019] [Indexed: 01/28/2023]
Abstract
The placenta is a transitory organ, located between the mother and the foetus, which supports intrauterine life. This organ has nutritional, endocrine and immunologic functions to support foetal development. Several factors are related to the correct functioning of the placenta including foetal and maternal blood flow, appropriate nutrients, expression and function of receptors and transporters, and the morphology of the placenta itself. Placental morphology is crucial for understanding the pathophysiology of the organ as represents the physical structure where nutrient exchange occurs. In pathologies of pregnancy such as diabetes mellitus in humans and animal models, several changes in the placental morphology occur, related mainly with placental size, hypervascularization, higher branching capillaries of the villi and increased glycogen deposits among others. Gestational diabetes mellitus is associated with modifications in the structure of the human placenta including changes in the surface area and volume, as well as histological changes including an increased volume of intervillous space and terminal villi, syncytiotrophoblast number, fibrinoid areas, and glycogen deposits. These modifications may result in functional changes in this organ thus limiting the wellbeing of the developing foetus. This review gives an overview of recurrent morphological changes at macroscopic and histological levels seen in the placenta from gestational diabetes in humans and animal models. This article is part of a Special Issue entitled: Membrane Transporters and Receptors in Pregnancy Metabolic Complications edited by Luis Sobrevia.
Collapse
|
49
|
Zhao S, Liao T, Zhou T, Huang X, Xiang H, Chen J, Xu Z. Formyl-peptide receptor 2 suppresses proliferation, migration and invasion in human extravillous trophoblastic cells. Prostaglandins Other Lipid Mediat 2019; 143:106342. [DOI: 10.1016/j.prostaglandins.2019.106342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 03/10/2019] [Accepted: 06/05/2019] [Indexed: 02/06/2023]
|
50
|
New Insights into the Process of Placentation and the Role of Oxidative Uterine Microenvironment. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9174521. [PMID: 31341539 PMCID: PMC6615000 DOI: 10.1155/2019/9174521] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/28/2019] [Indexed: 12/18/2022]
Abstract
For a successful pregnancy to occur, a predecidualized receptive endometrium must be invaded by placental differentiated cells (extravillous trophoblast cells (EVTs)) and, at the same time, continue decidualization. EVT invasion is aimed at anchoring the placenta to the maternal uterus and ensuring local blood supply increase necessary to provide normal placental and foetal development. The first is achieved by migrating through the maternal endometrium and deeper into the myometrium, while the second by transforming uterine spiral arteries into large vessels. This process is a tightly regulated battle comprising interests of both the mother and the foetus. Invading EVTs are required to perform a scope of functions: move, adhere, proliferate, differentiate, interact, and digest the extracellular matrix (ECM); tolerate hypoxia; transform the maternal spiral arteries; and die by apoptosis. All these functions are modulated by their surrounding microenvironment: oxygen, soluble factors (e.g., cytokines, growth factors, and hormones), ECM proteins, and reactive oxygen species. A deeper comprehension of oxidative uterine microenvironment contribution to trophoblast function will be addressed in this review.
Collapse
|