1
|
Zheng S, Feng W, Sun Z, Xu P, Dong S, Pan L, Shen H, He J, Chen P, Shu C. HSD17B1-mediated trophoblast differentiation lowers estrogen levels in early-onset preeclampsia. Sci Rep 2025; 15:17448. [PMID: 40394177 PMCID: PMC12092795 DOI: 10.1038/s41598-025-02490-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 05/13/2025] [Indexed: 05/22/2025] Open
Abstract
Early-onset preeclampsia (EOPE) with fetal growth restriction (FGR) is a severe hypertensive disorder of pregnancy characterized by placental dysfunction and estrogen deficiency. Based on single-cell RNA sequencing (scRNA-seq) profiling of specific placental trophoblast subtypes from EOPE-FGR and normotensive pregnancies, we identified HSD17B1, which encodes a key enzyme mediating estradiol conversion, as the central dysregulated node in EOPE pathogenesis. Multi-modal computational analysis (cluster annotation, cellular proportion calculation, comparison of differentially expressed genes, and characterization of cellular developmental trajectories) revealed key expression dynamics during syncytiotrophoblast (SCT) differentiation, with substantial suppression in EOPE specimens. Further validation using clinical placental samples confirmed the downregulation of HSD17B1 at the protein level in patients with EOPE, as demonstrated by immunohistochemistry and western blotting. Mechanistically, HSD17B1 knockdown in BeWo trophoblast models recapitulated the core EOPE phenotypes of impaired SCT differentiation and estrogen biosynthesis blockade. These findings reveal that HSD17B1 is a master coordinator of trophoblast-endocrine crosstalk, the impairment of which in placental trophoblasts may contribute to EOPE pathogenesis. Our findings provide a mechanistic basis for developing HSD17B1-targeted interventions that could contribute to the concurrent restoration of placental competence and hormonal regulation, improving the perinatal outcomes of patients with EOPE.
Collapse
Affiliation(s)
- Shu Zheng
- Department of Obstetrics, Obstetrics and Gynecology Center, The First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Wei Feng
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Zewen Sun
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Peng Xu
- Department of Obstetrics, Obstetrics and Gynecology Center, The First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Shuai Dong
- Department of Obstetrics, Obstetrics and Gynecology Center, The First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Lin Pan
- The First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Huimin Shen
- Department of Obstetrics, Obstetrics and Gynecology Center, The First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Jin He
- Department of Obstetrics, Obstetrics and Gynecology Center, The First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Peng Chen
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, China.
| | - Chang Shu
- Department of Obstetrics, Obstetrics and Gynecology Center, The First Hospital of Jilin University, Jilin University, Changchun, 130021, China.
| |
Collapse
|
2
|
Barbouti A, Varvarousis DN, Kanavaros P. The Role of Oxidative Stress-Induced Senescence in the Pathogenesis of Preeclampsia. Antioxidants (Basel) 2025; 14:529. [PMID: 40427411 PMCID: PMC12108173 DOI: 10.3390/antiox14050529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
Preeclampsia is a hypertension condition of human pregnancy that poses a significant risk to pregnant women and their fetus. It complicates about 2-8% of human pregnancies worldwide and displays multifactorial pathogenesis, including increased placental oxidative stress because of disturbed utero-placental blood flow. Recent evidence suggests that increased oxidative stress promotes acceleration of the placental senescence which is implicated in the pathogenesis of preeclampsia. This review focuses on the mechanisms that lead to oxidative stress in preeclamptic patients and examines the role of oxidative stress-induced placental senescence in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Alexandra Barbouti
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (D.N.V.); (P.K.)
| | | | | |
Collapse
|
3
|
Meinhardt G, Waldhäusl H, Lackner AI, Wächter J, Maxian T, Höbler AL, Vondra S, Kunihs V, Saleh L, Haslinger P, Kiraly P, Szilagyi A, Than NG, Pollheimer J, Haider S, Knöfler M. The multifaceted roles of the transcriptional coactivator TAZ in extravillous trophoblast development of the human placenta. Proc Natl Acad Sci U S A 2025; 122:e2426385122. [PMID: 40228123 PMCID: PMC12037006 DOI: 10.1073/pnas.2426385122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 03/07/2025] [Indexed: 04/16/2025] Open
Abstract
Insights into the molecular processes that drive early development of the human placenta is crucial for our understanding of pregnancy complications such as preeclampsia and fetal growth restriction, since defects in maturation of its epithelial cell, the trophoblast, have been detected in the severe forms of these diseases. However, key regulators specifying the differentiated trophoblast subtypes of the placenta are only slowly emerging. By using diverse trophoblast cell models, we herein show that the transcriptional coactivator of HIPPO signaling, TAZ, plays a pivotal role in the development of invasive extravillous trophoblasts (EVTs), cells that are essential for decidual vessel remodeling and adaption of maternal blood flow to the placenta. Ribonucleic acid sequencing (RNA-seq) or protein analyses upon TAZ gene silencing or CRISPR-Cas9-mediated knockout in differentiating trophoblast stem cells, organoids, primary EVTs, choriocarcinoma cells, or villous explant cultures unraveled that the coactivator promoted expression of genes associated with EVT identity, motility, and survival. Accordingly, depletion or chemical inhibition of TAZ, interacting with TEA domain family member 1 (TEAD1), impaired EVT differentiation, invasion, and migration and triggered apoptosis in the different trophoblast models. Notably, the coactivator also suppressed cell cycle genes and regulators of trophoblast self-renewal and prevented EVTs from cell fusion in organoids and primary cultures. Moreover, TAZ promoted human leukocyte antigen G (HLA-G) surface expression and increased NUAK1 kinase in EVTs thereby maintaining its own expression. In summary, the transcriptional coactivator TAZ plays a multifaceted role in the development of the EVT cell lineage by controlling different biological processes that initiate and preserve differentiation.
Collapse
Affiliation(s)
- Gudrun Meinhardt
- Placental Development Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Hanna Waldhäusl
- Placental Development Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Andreas I. Lackner
- Maternal-Fetal Immunology Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Jasmin Wächter
- Placental Development Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Theresa Maxian
- Placental Development Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Anna-Lena Höbler
- Maternal-Fetal Immunology Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Sigrid Vondra
- Maternal-Fetal Immunology Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Victoria Kunihs
- Placental Development Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Leila Saleh
- Placental Development Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Peter Haslinger
- Maternal-Fetal Immunology Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Peter Kiraly
- Systems Biology of Reproduction Lendulet Group, Institute of Molecular Life Sciences, Hungarian Research Network (HUN-REN) Research Centre for Natural Sciences, Budapest1117, Hungary
| | - Andras Szilagyi
- Systems Biology of Reproduction Lendulet Group, Institute of Molecular Life Sciences, Hungarian Research Network (HUN-REN) Research Centre for Natural Sciences, Budapest1117, Hungary
| | - Nandor G. Than
- Systems Biology of Reproduction Lendulet Group, Institute of Molecular Life Sciences, Hungarian Research Network (HUN-REN) Research Centre for Natural Sciences, Budapest1117, Hungary
- Maternity Private Clinic of Obstetrics and Gynecology, Budapest1126, Hungary
- Department of Obstetrics and Gynecology, Semmelweis University, Budapest1088, Hungary
| | - Jürgen Pollheimer
- Maternal-Fetal Immunology Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Sandra Haider
- Placental Development Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Martin Knöfler
- Placental Development Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| |
Collapse
|
4
|
Dimova T, Alexandrova M, Vangelov I, You Y, Mor G. The modeling of human implantation and early placentation: achievements and perspectives. Hum Reprod Update 2025; 31:133-163. [PMID: 39673726 DOI: 10.1093/humupd/dmae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 10/29/2024] [Indexed: 12/16/2024] Open
Abstract
BACKGROUND Successful implantation is a critical step for embryo survival. The major losses in natural and assisted human reproduction appeared to occur during the peri-implantation period. Because of ethical constraints, the fascinating maternal-fetal crosstalk during human implantation is difficult to study and thus, the possibility for clinical intervention is still limited. OBJECTIVE AND RATIONALE This review highlights some features of human implantation as a unique, ineffective and difficult-to-model process and summarizes the pros and cons of the most used in vivo, ex vivo and in vitro models. We point out the variety of cell line-derived models and how these data are corroborated by well-defined primary cells of the same nature. Important aspects related to the handling, standardization, validation, and modus operandi of the advanced 3D in vitro models are widely discussed. Special attention is paid to blastocyst-like models recapitulating the hybrid phenotype and HLA profile of extravillous trophoblasts, which are a unique yet poorly understood population with a major role in the successful implantation and immune mother-embryo recognition. Despite raising new ethical dilemmas, extended embryo cultures and synthetic embryo models are also in the scope of our review. SEARCH METHODS We searched the electronic database PubMed from inception until March 2024 by using a multi-stage search strategy of MeSH terms and keywords. In addition, we conducted a forward and backward reference search of authors mentioned in selected articles. OUTCOMES Primates and rodents are valuable in vivo models for human implantation research. However, the deep interstitial, glandular, and endovascular invasion accompanied by a range of human-specific factors responsible for the survival of the fetus determines the uniqueness of the human implantation and limits the cross-species extrapolation of the data. The ex vivo models are short-term cultures, not relevant to the period of implantation, and difficult to standardize. Moreover, the access to tissues from elective terminations of pregnancy raises ethical and legal concerns. Easy-to-culture cancer cell lines have many limitations such as being prone to spontaneous transformation and lacking decent tissue characteristics. The replacement of the original human explants, primary cells or cancer cell lines with cultures of immortalized cell lines with preserved stem cell characteristics appears to be superior for in vitro modeling of human implantation and early placentation. Remarkable advances in our understanding of the peri-implantation stages have also been made by advanced three dimensional (3D) models i.e. spheroids, organoids, and assembloids, as placental and endometrial surrogates. Much work remains to be done for the optimization and standardization of these integrated and complex models. The inclusion of immune components in these models would be an asset to delineate mechanisms of immune tolerance. Stem cell-based embryo-like models and surplus IVF embryos for research bring intriguing possibilities and are thought to be the trend for the next decade for in vitro modeling of human implantation and early embryogenesis. Along with this research, new ethical dilemmas such as the moral status of the human embryo and the potential exploitation of women consenting to donate their spare embryos have emerged. The careful appraisal and development of national legal and ethical frameworks are crucial for better regulation of studies using human embryos and embryoids to reach the potential benefits for human reproduction. WIDER IMPLICATIONS We believe that our data provide a systematization of the available information on the modeling of human implantation and early placentation and will facilitate further research in this field. A strict classification of the advanced 3D models with their pros, cons, applicability, and availability would help improve the research quality to provide reliable outputs.
Collapse
Affiliation(s)
- Tanya Dimova
- Institute of Biology and Immunology of Reproduction "Acad. Kiril Bratanov", Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Marina Alexandrova
- Institute of Biology and Immunology of Reproduction "Acad. Kiril Bratanov", Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Ivaylo Vangelov
- Institute of Biology and Immunology of Reproduction "Acad. Kiril Bratanov", Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Yuan You
- C.S. Mott Center for Human Growth and Development, Wayne State University, Detroit, MI, USA
| | - Gil Mor
- C.S. Mott Center for Human Growth and Development, Wayne State University, Detroit, MI, USA
| |
Collapse
|
5
|
Di Giorgio E, Xodo S, Orsaria M, Mariuzzi L, Picco R, Tolotto V, Cortolezzis Y, D'Este F, Grandi N, Driul L, Londero A, Xodo LE. The central role of creatine and polyamines in fetal growth restriction. FASEB J 2024; 38:e70222. [PMID: 39614665 DOI: 10.1096/fj.202401946r] [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: 08/20/2024] [Revised: 10/23/2024] [Accepted: 11/19/2024] [Indexed: 12/01/2024]
Abstract
Placental insufficiency often correlates with fetal growth restriction (FGR), a condition that has both short- and long-term effects on the health of the newborn. In our study, we analyzed placental tissue from infants with FGR and from infants classified as small for gestational age (SGA) or appropriate for gestational age (AGA), performing comprehensive analyses that included transcriptomics and metabolomics. By examining villus tissue biopsies and 3D trophoblast organoids, we identified significant metabolic changes in placentas associated with FGR. These changes include adaptations to reduced oxygen levels and modifications in arginine metabolism, particularly within the polyamine and creatine phosphate synthesis pathways. Specifically, we found that placentas with FGR utilize arginine to produce phosphocreatine, a crucial energy reservoir for ATP production that is essential for maintaining trophoblast function. In addition, we found polyamine insufficiency in FGR placentas due to increased SAT1 expression. SAT1 facilitates the acetylation and subsequent elimination of spermine and spermidine from trophoblasts, resulting in a deficit of polyamines that cannot be compensated by arginine or polyamine supplementation alone, unless SAT1 expression is suppressed. Our study contributes significantly to the understanding of metabolic adaptations associated with placental dysfunction and provides valuable insights into potential therapeutic opportunities for the future.
Collapse
Affiliation(s)
| | - Serena Xodo
- Clinic of Obstetrics and Gynecology, Santa Maria della Misericordia Hospital, ASUFC, Udine, Italy
| | - Maria Orsaria
- Institute of Pathology, Department of Medicine, University of Udine, Udine, Italy
| | - Laura Mariuzzi
- Institute of Pathology, Department of Medicine, University of Udine, Udine, Italy
| | | | | | | | | | - Nicole Grandi
- Laboratory of Molecular Virology, Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Lorenza Driul
- Department of Medicine, University of Udine, Udine, Italy
- Clinic of Obstetrics and Gynecology, Santa Maria della Misericordia Hospital, ASUFC, Udine, Italy
| | - Ambrogio Londero
- Obstetrics and Gynecology Unit, IRCCS Institute Giannina Gaslini, Genova, Italy
| | - Luigi E Xodo
- Department of Medicine, University of Udine, Udine, Italy
| |
Collapse
|
6
|
Li Y, Ma L, He R, Teng F, Qin X, Liang X, Wang J. Pregnancy Metabolic Adaptation and Changes in Placental Metabolism in Preeclampsia. Geburtshilfe Frauenheilkd 2024; 84:1033-1042. [PMID: 39524034 PMCID: PMC11543110 DOI: 10.1055/a-2403-4855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/24/2024] [Indexed: 11/16/2024] Open
Abstract
Pregnancy is a unique physiological state in which the maternal body undergoes a series of changes in the metabolism of glucose, lipids, amino acids, and other nutrients in order to adapt to the altered state of pregnancy and provide adequate nutrients for the fetus' growth and development. The metabolism of various nutrients is regulated by one another in order to maintain homeostasis in the body. Failure to adapt to the altered physiological conditions of pregnancy can lead to a range of pregnancy issues, including fetal growth limitation and preeclampsia. A failure of metabolic adaptation during pregnancy is linked to the emergence of preeclampsia. The treatment of preeclampsia by focusing on metabolic changes may provide new therapeutic alternatives.
Collapse
Affiliation(s)
- Yaxi Li
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Ling Ma
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Ruifen He
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Fei Teng
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Xue Qin
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Xiaolei Liang
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Key Laboratory for Gynecologic Oncology Gansu Province, Lanzhou, China
| | - Jing Wang
- The First Clinical Medical College of Lanzhou University, the First Hospital of Lanzhou University, Lanzhou City, Gansu Province, China
| |
Collapse
|
7
|
Siriwardena D, Munger C, Penfold C, Kohler TN, Weberling A, Linneberg-Agerholm M, Slatery E, Ellermann AL, Bergmann S, Clark SJ, Rawlings TM, Brickman JM, Reik W, Brosens JJ, Zernicka-Goetz M, Sasaki E, Behr R, Hollfelder F, Boroviak TE. Marmoset and human trophoblast stem cells differ in signaling requirements and recapitulate divergent modes of trophoblast invasion. Cell Stem Cell 2024; 31:1427-1446.e8. [PMID: 39321797 PMCID: PMC7616712 DOI: 10.1016/j.stem.2024.09.004] [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: 03/13/2023] [Revised: 06/01/2024] [Accepted: 09/05/2024] [Indexed: 09/27/2024]
Abstract
Early human trophoblast development has remained elusive due to the inaccessibility of the early conceptus. Non-human primate models recapitulate many features of human development and allow access to early postimplantation stages. Here, we tracked the pre- to postimplantation transition of the trophoblast lineage in superficially implanting marmoset embryos in vivo. We differentiated marmoset naive pluripotent stem cells into trophoblast stem cells (TSCs), which exhibited trophoblast-specific transcriptome, methylome, differentiation potential, and long-term self-renewal. Notably, human TSC culture conditions failed to support marmoset TSC derivation, instead inducing an extraembryonic mesoderm-like fate in marmoset cells. We show that combined MEK, TGF-β/NODAL, and histone deacetylase inhibition stabilizes a periimplantation trophoblast-like identity in marmoset TSCs. By contrast, these conditions differentiated human TSCs toward extravillous trophoblasts. Our work presents a paradigm to harness the evolutionary divergence in implantation strategies to elucidate human trophoblast development and invasion.
Collapse
Affiliation(s)
- Dylan Siriwardena
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge, UK; Wellcome Trust, Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Clara Munger
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge, UK; Wellcome Trust, Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK; Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Christopher Penfold
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge, UK; Wellcome Trust, Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Timo N Kohler
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | | | - Erin Slatery
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Anna L Ellermann
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Sophie Bergmann
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge, UK; Wellcome Trust, Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Stephen J Clark
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK; Altos Labs Cambridge Institute, Cambridge, UK; Epigenetics Programme, Babraham Institute, Cambridge, UK
| | - Thomas M Rawlings
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Joshua M Brickman
- Novo Nordisk Foundation Center for Stem Cell Medicine (renew), University of Copenhagen, Copenhagen, Denmark
| | - Wolf Reik
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK; Altos Labs Cambridge Institute, Cambridge, UK; Epigenetics Programme, Babraham Institute, Cambridge, UK; Wellcome Trust Sanger Institute, Cambridge, UK
| | - Jan J Brosens
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK; Tommy's National Centre for Miscarriage Research, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK
| | - Magdalena Zernicka-Goetz
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge, UK; Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Erika Sasaki
- Department of Marmoset Biology and Medicine, Central Institute for Experimental Animals, Kawasaki 210-0821, Japan
| | - Rüdiger Behr
- German Primate Center, Leibniz-Institute for Primate Research, Göttingen, Germany; DZHK (German Center for Cardiovascular Research), Göttingen, Germany
| | | | - Thorsten E Boroviak
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge, UK; Wellcome Trust, Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
8
|
Balestrini PA, Abdelbaki A, McCarthy A, Devito L, Senner CE, Chen AE, Munusamy P, Blakeley P, Elder K, Snell P, Christie L, Serhal P, Odia RA, Sangrithi M, Niakan KK, Fogarty NME. Transcription factor-based transdifferentiation of human embryonic to trophoblast stem cells. Development 2024; 151:dev202778. [PMID: 39250534 PMCID: PMC11556314 DOI: 10.1242/dev.202778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 08/05/2024] [Indexed: 09/11/2024]
Abstract
During the first week of development, human embryos form a blastocyst composed of an inner cell mass and trophectoderm (TE) cells, the latter of which are progenitors of placental trophoblast. Here, we investigated the expression of transcripts in the human TE from early to late blastocyst stages. We identified enrichment of the transcription factors GATA2, GATA3, TFAP2C and KLF5 and characterised their protein expression dynamics across TE development. By inducible overexpression and mRNA transfection, we determined that these factors, together with MYC, are sufficient to establish induced trophoblast stem cells (iTSCs) from primed human embryonic stem cells. These iTSCs self-renew and recapitulate morphological characteristics, gene expression profiles, and directed differentiation potential, similar to existing human TSCs. Systematic omission of each, or combinations of factors, revealed the crucial importance of GATA2 and GATA3 for iTSC transdifferentiation. Altogether, these findings provide insights into the transcription factor network that may be operational in the human TE and broaden the methods for establishing cellular models of early human placental progenitor cells, which may be useful in the future to model placental-associated diseases.
Collapse
Affiliation(s)
- Paula A. Balestrini
- Centre for Gene Therapy and Regenerative Medicine, King's College London, London SE1 9RT, UK
| | - Ahmed Abdelbaki
- Human Embryo and Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- The Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
- Department of Zoology, Faculty of Science, Zagazig University, Zagazig 44519, Egypt
| | - Afshan McCarthy
- Human Embryo and Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Liani Devito
- Human Embryo and Stem Cell Unit, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Claire E. Senner
- The Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Alice E. Chen
- Trestle Biotherapeutics, Centre for Novel Therapeutics, 9310 Athena Circle, La Jolla, CA 92037, USA
| | - Prabhakaran Munusamy
- KK Women's and Children's Hospital, Division of Obstetrics and Gynecology, 100 Bukit Timah Road, Singapore229899, Singapore
| | - Paul Blakeley
- Human Embryo and Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Department of Surgery, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kay Elder
- Bourn Hall Clinic, Bourn, Cambridge CB23 2TN, UK
| | - Phil Snell
- Bourn Hall Clinic, Bourn, Cambridge CB23 2TN, UK
| | | | - Paul Serhal
- The Centre for Reproductive & Genetic Health, 230–232 Great Portland Street, London W1W 5QS, UK
| | - Rabi A. Odia
- The Centre for Reproductive & Genetic Health, 230–232 Great Portland Street, London W1W 5QS, UK
| | - Mahesh Sangrithi
- Centre for Gene Therapy and Regenerative Medicine, King's College London, London SE1 9RT, UK
- KK Women's and Children's Hospital, Division of Obstetrics and Gynecology, 100 Bukit Timah Road, Singapore229899, Singapore
- Duke-NUS Graduate Medical School, Cancer Stem Cell Biology/OBGYN ACP, 8 College Road, Singapore 169857, Singapore
| | - Kathy K. Niakan
- Human Embryo and Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- The Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, 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
- Epigenetics Programme, Babraham Institute, Cambridge CB22 3AT, UK
| | - Norah M. E. Fogarty
- Centre for Gene Therapy and Regenerative Medicine, King's College London, London SE1 9RT, UK
- Human Embryo and Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| |
Collapse
|
9
|
Gupta A, Patel DJ, Pajai S. Exploring the Spectrum of Electrolyte Imbalances in Preeclampsia: Mechanisms, Implications, and Clinical Insights. Cureus 2024; 16:e67666. [PMID: 39314616 PMCID: PMC11418792 DOI: 10.7759/cureus.67666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 08/24/2024] [Indexed: 09/25/2024] Open
Abstract
Preeclampsia, a complex and perplexing disorder unique to pregnancy, is widely recognized as primarily originating from placental dysfunction and can only be resolved by the delivery of the fetus in severe cases. Preeclampsia is a prevalent medical issue during pregnancy and is associated with elevated rates of maternal and infant mortality and morbidity. The exact cause of preeclampsia remains uncertain, although multiple factors have been implicated in its development based on current knowledge. Preeclampsia is characterized by maternal endothelial dysfunction due to the presence of fetal-derived circulatory substances from the placenta. The condition is associated with various risk factors, including maternal comorbidities such as chronic renal disease, hypertension (HTN), and obesity. Additionally, a family history of preeclampsia, nulliparity, multiple gestations, previous instances of preeclampsia, or intrauterine fetal growth restriction (IUGR) are considered risk factors. Electrolytes, including sodium, potassium, and chloride, play a critical role in the function of vascular smooth muscles and may potentially contribute to the pathophysiology of hypertension. In this review, we have summarized the literature on electrolytes in preeclampsia by conducting an extensive systematic search of databases such as PubMed, Excerpta Medica database (EMBASE), and Medical Literature Analysis and Retrieval System Online (MEDLINE).
Collapse
Affiliation(s)
- Aishwarya Gupta
- Department of Obstetrics and Gynaecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Dharmesh J Patel
- Department of Obstetrics and Gynaecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sandhya Pajai
- Department of Obstetrics and Gynaecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
10
|
Park S, Hunter ES. Modeling the human placenta: in vitro applications in developmental and reproductive toxicology. Crit Rev Toxicol 2024; 54:431-464. [PMID: 39016688 DOI: 10.1080/10408444.2023.2295349] [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: 07/31/2023] [Revised: 11/20/2023] [Accepted: 12/07/2023] [Indexed: 07/18/2024]
Abstract
During its temporary tenure, the placenta has extensive and specialized functions that are critical for pre- and post-natal development. The consequences of chemical exposure in utero can have profound effects on the structure and function of pregnancy-associated tissues and the life-long health of the birthing person and their offspring. However, the toxicological importance and critical functions of the placenta to embryonic and fetal development and maturation have been understudied. This narrative will review early placental development in humans and highlight some in vitro models currently in use that are or can be applied to better understand placental processes underlying developmental toxicity due to in utero environmental exposures.
Collapse
Affiliation(s)
- Sarah Park
- Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
- Center for Computational Toxicology and Exposure, ORD, US EPA, Research Triangle Park, NC, USA
| | - Edward Sidney Hunter
- Center for Computational Toxicology and Exposure, ORD, US EPA, Research Triangle Park, NC, USA
| |
Collapse
|
11
|
Bacon SJ, Zhu Y, Ghosh P. Early spiral arteriole remodeling in the uterine-placental interface: A rat model. J Anat 2024; 244:1054-1066. [PMID: 38288680 PMCID: PMC11095304 DOI: 10.1111/joa.14019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/12/2024] [Accepted: 01/12/2024] [Indexed: 05/16/2024] Open
Abstract
The mammalian placenta's interface with the parent is a richly vascularized tissue whose development relies upon communication between many different cell types within the uterine microenvironment. The uterine blood vessels of the interface are reshaped during pregnancy into wide-bore, flaccid vessels that convey parental blood to the exchange region of the placenta. Invasive trophoblast as well as parental uterine macrophages and Natural Killer cells are involved in the stepwise remodeling of these vessels and their respective contributions to this crucial process are still being delineated. However, the earliest steps in arteriole remodeling are understudied as they are difficult to study in humans, and other species lack the deep trophoblast invasion that is so prominent a feature of placentation in humans. Here, we further characterize the rat, with deep hemochorial placentation akin to humans, as a model system in which to tease apart the earliest, relatively understudied events in spiral arteriole remodeling. We show that the rat uterine-placental interface increases in size and vascularity rapidly, before trophoblast invasion. The remodeling stages in the arterioles of the rat uterine-placental interface follow a sequence of anatomical changes similar to those in humans, and there are changes to the arterioles' muscular tunica media prior to the marked influx of immune cells. The rat is a tractable model in which to better understand the cell/cell interactions occurring in vivo in an intact tissue microenvironment over time.
Collapse
Affiliation(s)
- Sarah J Bacon
- Department of Biological Sciences, Mount Holyoke College, South Hadley, Massachusetts, USA
| | - Yuxi Zhu
- Department of Biological Sciences, Mount Holyoke College, South Hadley, Massachusetts, USA
| | - Priyanjali Ghosh
- Department of Biological Sciences, Mount Holyoke College, South Hadley, Massachusetts, USA
| |
Collapse
|
12
|
Hoo R, Ruiz-Morales ER, Kelava I, Rawat M, Mazzeo CI, Tuck E, Sancho-Serra C, Chelaghma S, Predeus AV, Murray S, Fernandez-Antoran D, Waller RF, Álvarez-Errico D, Lee MCS, Vento-Tormo R. Acute response to pathogens in the early human placenta at single-cell resolution. Cell Syst 2024; 15:425-444.e9. [PMID: 38703772 DOI: 10.1016/j.cels.2024.04.002] [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: 05/19/2023] [Revised: 12/01/2023] [Accepted: 04/16/2024] [Indexed: 05/06/2024]
Abstract
The placenta is a selective maternal-fetal barrier that provides nourishment and protection from infections. However, certain pathogens can attach to and even cross the placenta, causing pregnancy complications with potential lifelong impacts on the child's health. Here, we profiled at the single-cell level the placental responses to three pathogens associated with intrauterine complications-Plasmodium falciparum, Listeria monocytogenes, and Toxoplasma gondii. We found that upon exposure to the pathogens, all placental lineages trigger inflammatory responses that may compromise placental function. Additionally, we characterized the responses of fetal macrophages known as Hofbauer cells (HBCs) to each pathogen and propose that they are the probable niche for T. gondii. Finally, we revealed how P. falciparum adapts to the placental microenvironment by modulating protein export into the host erythrocyte and nutrient uptake pathways. Altogether, we have defined the cellular networks and signaling pathways mediating acute placental inflammatory responses that could contribute to pregnancy complications.
Collapse
Affiliation(s)
- Regina Hoo
- Wellcome Sanger Institute, Cambridge, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | | | - Iva Kelava
- Wellcome Sanger Institute, Cambridge, UK
| | - Mukul Rawat
- Wellcome Sanger Institute, Cambridge, UK; Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee, UK
| | | | | | | | - Sara Chelaghma
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | | | - David Fernandez-Antoran
- Wellcome/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge, UK; Department of Pathology, University of Cambridge, Cambridge, UK
| | - Ross F Waller
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | - Marcus C S Lee
- Wellcome Sanger Institute, Cambridge, UK; Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, University of Dundee, Dundee, UK.
| | - Roser Vento-Tormo
- Wellcome Sanger Institute, Cambridge, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| |
Collapse
|
13
|
Lyssy F, Forstner D, Brugger BA, Ujčič K, Guettler J, Kupper N, Wernitznig S, Daxboeck C, Neuper L, El-Heliebi A, Kloimboeck T, Kargl J, Huppertz B, Ghaffari-Tabrizi-Wizsy N, Gauster M. The chicken chorioallantoic membrane assay revisited - A face-lifted approach for new perspectives in placenta research. Placenta 2024:S0143-4004(24)00113-9. [PMID: 38705802 DOI: 10.1016/j.placenta.2024.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/09/2024] [Accepted: 04/29/2024] [Indexed: 05/07/2024]
Abstract
The study of very early human placentation is largely limited due to ethical restrictions on the use of embryonic tissue and the fact that the placental anatomy of common laboratory animal models varies considerably from that of humans. In recent years several promising models, including trophoblast stem cell-derived organoids, have been developed that have also proven useful for the study of important trophoblast differentiation processes. However, the consideration of maternal blood flow in trophoblast invasion models currently appears to be limited to animal models. An almost forgotten model to study the invasive behavior of trophoblasts is to culture them in vitro on the chicken chorioallantoic membrane (CAM), showing an extraembryonic vascular network in its mesenchymal stroma that is continuously perfused by the chicken embryonic blood circulation. Here, we present an extension of the previously described ex ovo CAM assay and describe the use of cavity-bearing trophoblast spheroids obtained from the first trimester cell line ACH-3P. We demonstrate how spheroids penetrated the CAM and that erosion of CAM vessels by trophoblasts led to filling of the spheroid cavities with chicken blood, mimicking initial steps of intervillous space blood perfusion. Moreover, we prove that this model is useful for state-of-the-art techniques including immunofluorescence and in situ padlock probe hybridization, making it a versatile tool to study aspects of trophoblast invasion in presence of blood flow.
Collapse
Affiliation(s)
- Freya Lyssy
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Désirée Forstner
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Beatrice A Brugger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Kaja Ujčič
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Jacqueline Guettler
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Nadja Kupper
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Stefan Wernitznig
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Christine Daxboeck
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Lena Neuper
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Amin El-Heliebi
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Teresa Kloimboeck
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Austria
| | - Julia Kargl
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Austria
| | - Berthold Huppertz
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | | | - Martin Gauster
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria.
| |
Collapse
|
14
|
Ding Z, Wu L, Sun Y, Zhu Y, Zuo Q, Yuan L, Wang C, Sun L, Xu Y, Zhang Y. Downregulated PDIA3P1 lncRNA Impairs Trophoblast Phenotype by Regulating Snail and SFRP1 in PE. Anal Cell Pathol (Amst) 2024; 2024:8972022. [PMID: 38715918 PMCID: PMC11074859 DOI: 10.1155/2024/8972022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/11/2024] [Accepted: 03/02/2024] [Indexed: 06/04/2024] Open
Abstract
Preeclampsia (PE) manifests as a pregnancy-specific complication arising from compromised placentation characterized by inadequate trophoblast invasion. A growing body of evidence underscores the pivotal involvement of pseudogenes, a subset of long noncoding RNAs, in the pathological processes of PE. This study presents a novel finding, demonstrating a significant downregulation of the pseudogene PDIA3P1 in PE placental tissues compared to normal tissues. In vitro functional assays revealed that suppressing PDIA3P1 hindered trophoblast proliferation, invasion, and migration, concurrently upregulating the expression of secreted frizzled-related protein 1 (SFRP1). Further exploration of the regulatory role of PDIA3P1 in PE, utilizing human trophoblasts, established that PDIA3P1 exerts its function by binding to HuR, thereby enhancing the stability of Snail expression in trophoblasts. Overall, our findings suggest a crucial role for PDIA3P1 in regulating trophoblast properties and contributing to the pathogenesis of PE, offering potential targets for prognosis and therapeutic intervention.
Collapse
Affiliation(s)
- Zhengzheng Ding
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
- Department of Obstetrics and Gynecology, Nanjing Maternity and Child Health Care Hospital, Women' s Hospital of Nanjing Medical University, 123 Tianfeixiang, Mochou Road, Qinhuai District, Nanjing 210004, Jiangsu, China
| | - Liuxin Wu
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Yue Sun
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Yuanyuan Zhu
- Department of Obstetrics and Gynecology, Nanjing Maternity and Child Health Care Hospital, Women' s Hospital of Nanjing Medical University, 123 Tianfeixiang, Mochou Road, Qinhuai District, Nanjing 210004, Jiangsu, China
| | - Qing Zuo
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Li Yuan
- Department of Obstetrics and Gynecology, Taizhou Maternity Hospital Affiliated to Nantong University, Taizhou 225300, Jiangsu, China
| | - Cong Wang
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Lizhou Sun
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Yetao Xu
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Yuanyuan Zhang
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| |
Collapse
|
15
|
Hua Q, Li Z, Zhou Y, Wang Y, Yu Y, Sun L, Ye J, Li L. Single-cell RNA sequencing reveals association of aberrant placental trophoblasts and FN1 reduction in late-onset fetal growth restriction. Placenta 2024; 146:30-41. [PMID: 38160601 DOI: 10.1016/j.placenta.2023.12.022] [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: 09/14/2023] [Revised: 12/06/2023] [Accepted: 12/19/2023] [Indexed: 01/03/2024]
Abstract
INTRODUCTION Fetal growth restriction (FGR) can lead to fetal mental development abnormalities, malformations, and even intrauterine death. Defects in the trophoblasts at the maternal-fetal interface may contribute to FGR. However, the impact of trophoblasts on FGR is still not well understood. Therefore, the objective of this study is to characterize the heterogeneity of placental cells at the single-cell level and investigate the role of trophoblast subtypes in the pathogenesis of FGR at the cellular and molecular levels. METHODS Single-cell RNA sequencing was performed on the maternal side of placentas from two normal pregnant women and two pregnant women with FGR. Lentivirus transfection was used to establish a FN1 knockout model in trophoblast HTR-8-Svneo cells. The effect of FN1 knockout on cell migration and invasion of HTR-8-Svneo cells was assessed through wound healing and transwell assays. RESULTS Nine cell types were annotated in 39,161 cells derived from single-cell RNA sequencing. The FGR group exhibited a decrease in the percentage of trophoblasts, especially in subtype of extravillous trophoblasts (EVTs). The expression of FN1 was reduced in trophoblasts and EVTs. Furthermore, the protein expression levels of FN1 in the placentas of FGR patients were significantly lower than those of normal pregnant women. The cell migration and invasion ability of HTR-8-Svneo cells were inhibited after the knockdown of FN1. DISCUSSION The dysregulation of the trophoblast subtype-EVTs is involved in placental dysplasia related to FGR. The association between aberrant placental trophoblasts and reduced FN1 expression may contribute to insufficient remodeling of spiral arteries and the formation of FGR.
Collapse
Affiliation(s)
- Qing Hua
- Department of Obstetrics and Gynecology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007, Henan, PR China
| | - Zhe Li
- Department of Obstetrics and Gynecology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007, Henan, PR China
| | - Yadan Zhou
- Department of Obstetrics and Gynecology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007, Henan, PR China
| | - Yali Wang
- Department of Obstetrics and Gynecology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007, Henan, PR China
| | - Yangyang Yu
- Department of Obstetrics and Gynecology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007, Henan, PR China
| | - Lei Sun
- Stem Cell Regenerative Medicine Transformation Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007, Henan, PR China
| | - Jianping Ye
- Metabolic Disease Research Center, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007, PR China; Center for Advanced Medicine, College of Medicine, Zhengzhou University, Zhengzhou, 450007, Henan, PR China.
| | - Li Li
- Department of Obstetrics and Gynecology, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007, Henan, PR China.
| |
Collapse
|
16
|
Jia X, Yang S, Wang X, Ruan J, Huang W. HOXB3 promotes trophoblast cell proliferation, invasion, and migration to alleviate preeclampsia via mediating the Notch/Wnt/β-catenin pathway. Eur J Pharmacol 2023; 960:176015. [PMID: 37652291 DOI: 10.1016/j.ejphar.2023.176015] [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: 11/22/2022] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/02/2023]
Abstract
Preeclampsia (PE) harms a significant number of pregnant women and fetuses. However, because of its complex pathological mechanisms, there is no cure except for delivery. This study identified the impact and mechanisms of action of HOXB3 in PE. The behaviors of HTR-8/SVneo cells were analyzed using a cell counting kit-8, EdU, and transwell assays. The interaction between HOXB3 and Notch1 was assessed using a luciferase reporter and chromatin immunoprecipitation assays. Expression was measured by quantitative real-time polymerase chain reaction, western blotting, and immunofluorescence assays. Additionally, the function of HOXB3 was evaluated in an established rat model of PE. We found that HOXB3 was upregulated in PE. HOXB3 overexpression facilitated trophoblast cell proliferation, migration, and invasion. HOXB3 transcriptionally regulated Notch1 by binding to its promoter. Notch1 knockdown abrogated the functions of HOXB3 and the-catenin pathway in trophoblasts. Suppression of the Wnt/β-catenin pathway abrogated the effects of HOXB3. Additionally, HOXB3 alleviated the symptoms in PE rats. In conclusion, HOXB3 transcriptionally activated Notch1 expression and the-catenin pathway, promoting trophoblast cell proliferation, invasion, and migration, thereby alleviating PE progression. This study provides a novel approach for PE therapy.
Collapse
Affiliation(s)
- Xueqin Jia
- Department of Obstetrics, Xinhui People's Hospital of Southern Medical University, Jiangmen, 529100, Guangdong, China; Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China; Department of Obstetrics, The People's Hospital of Rizhao, Rizhao, Shandong, 276800, China
| | - Shiying Yang
- Department of Obstetrics, The People's Hospital of Rizhao, Rizhao, Shandong, 276800, China
| | - Xia Wang
- Department of Obstetrics, The People's Hospital of Rizhao, Rizhao, Shandong, 276800, China
| | - Jianbing Ruan
- Department of Obstetrics, Xinhui People's Hospital of Southern Medical University, Jiangmen, 529100, Guangdong, China.
| | - Wenhua Huang
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Digital Medicine and Biomechanics, National Key Discipline of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
17
|
Yokouchi-Konishi T, Liu Y, Feng L. Progesterone receptor membrane component 2 is critical for human placental extravillous trophoblast invasion. Biol Reprod 2023; 109:759-771. [PMID: 37665239 DOI: 10.1093/biolre/ioad109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 08/14/2023] [Accepted: 08/30/2023] [Indexed: 09/05/2023] Open
Abstract
Proper extravillous trophoblast invasion is essential for normal placentation and pregnancy. However, the molecular mechanisms by which cytotrophoblasts differentiate into extravillous trophoblast are unclear. We discovered that in the first-trimester placenta, progesterone receptor membrane component 2 was highly expressed in syncytiotrophoblast but significantly lower in extravillous trophoblast and cytotrophoblasts, indicating a divergent role for progesterone receptor membrane component 2 in trophoblast functions. We aim to examine the role of progesterone receptor membrane component 2 in extravillous trophoblasts invasion mediated by both intracellular and extracellular signals. Progesterone receptor membrane component 2 knockdown and overexpression cells were established in HTR8/SVneo cells, a first-trimester extravillous trophoblast-derived cell model, by transfection with small-interfering RNA or progesterone receptor membrane component 2 plasmids, respectively. Progesterone receptor membrane component 2 knockdown led to cellular morphological changes , enhanced trophoblast proliferation,invasion, and promoted tube formation. These effects were mediated by the activation of hypoxia-inducible factor 1alpha and an increased expression of vascular endothelial growth factor A. The culture supernatant collected from progesterone receptor membrane component 2 knockdown cells did not significantly affect extravillous trophoblast invasion compared to the controls, indicating that extracellular signaling did not robustly regulate extravillous trophoblast invasion in this study. In conclusion, attenuation of progesterone receptor membrane component 2 plays a role in placentation by promoting cell proliferation, invasion, and angiogenesis in extravillous trophoblasts via activation of hypoxia-inducible factor 1 alpha signaling. We thus identified a new function of progesterone receptor membrane component 2 and provide insights on understanding the mechanisms of trophoblast invasion.
Collapse
Affiliation(s)
- Tae Yokouchi-Konishi
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, USA
- Department of Obstetrics and Gynecology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Yongjie Liu
- Ministry of Education and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liping Feng
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
18
|
Zhang S, Shi Y, Dong P. USP8 targeted by Mir-874-3p promotes trophoblastic cell invasion by stabilizing the expression of ENaC on trophoblast membrane. Hum Immunol 2023; 84:618-630. [PMID: 37741774 DOI: 10.1016/j.humimm.2023.09.001] [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: 08/04/2022] [Revised: 07/21/2023] [Accepted: 09/05/2023] [Indexed: 09/25/2023]
Abstract
The aim of this study was to investigate the role of ubiquitin-specific peptidase 8 (USP8) in human trophoblast cells and its molecular mechanism. Based on the GSE30186 dataset, USP8 was identified as a downregulated gene in pre-eclampsia (PE). Analysis of clinical samples also revealed that USP8 expression at both the mRNA and protein levels in placental tissue from patients with PE was significantly lower than that from healthy pregnant women. Plate clone formation, scratch-wound healing, Transwell, tubule formation, and western blot assays collectively revealed that USP8 overexpression promoted the proliferation, migration, invasion, and pro-angiogenesis function of trophoblast cells, while USP8 knockdown induced the opposite effects. Bioinformatics analysis and luciferase reporter assay results indicated that the 3' untranslated region of USP8 was targeted by miR-874-3p. USP8 expression in the placental tissue of patients with PE was significantly lower than that of healthy pregnant women. USP8 actively regulated the growth and invasion of human trophoblast cells and stabilized the epithelial sodium channel (ENaC) on the cell membrane. MiR-874 targeted USP8 in the trophoblast cells and upregulation of miR-874-3p resulted in a decrease in the proliferation, migration, invasion, and pro-angiogenesis ability of trophoblast cells. These results indicate that USP8 can reverse the above mentioned negative effects of miR-874-3p on trophoblast cells. USP8 targeted by miR-874-3p facilitates the invasion of trophoblastic cells by stabilizing the expression of the ENaC, which may be a possible therapeutic target for PE.
Collapse
Affiliation(s)
- Suqin Zhang
- Department of Maternity, Yantaishan Hospital, Yantai, Shandong, China.
| | - Yanmei Shi
- Department of Maternity, Yantaishan Hospital, Yantai, Shandong, China.
| | - Pingping Dong
- Department of Maternity, Yantaishan Hospital, Yantai, Shandong, China.
| |
Collapse
|
19
|
Gupta SK, Mochan S, Arora P, Rani N, Luthra K, Dwivedi S, Bhatla N, Kshetrapal P, Dhingra R. Hydrogen sulfide promotes migration of trophoblast cells by a Rho GTPase mediated actin cytoskeleton reorganization. Placenta 2023; 142:135-146. [PMID: 37774537 DOI: 10.1016/j.placenta.2023.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/24/2023] [Accepted: 09/07/2023] [Indexed: 10/01/2023]
Abstract
INTRODUCTION Preeclampsia (PE) arises due to defective spiral artery remodelling which may be due to deficient migration of trophoblast cells. Migration of human endothelial cells has been shown to be promoted via Hydrogen sulphide(H2S)/Rho GTPase Rac1 axis. This novel role of H2S and its downstream processes have not yet been studied in the development and function of the placental trophoblast cells. METHODS Placental tissues were obtained post-delivery from consented preeclamptic and normotensive mothers (n = 60). The protein expression levels of cystathionine-gamma-lyase (CSE) and cystathionine-beta-synthase (CBS) along with its downstream migratory molecules were compared in both the arms. The pro-migratory role of H2S was investigated in a first trimester placental cell line. RESULTS H2S promoted the migration of trophoblast cells in a Rho GTPase dependent manner mediated by actin cytoskeleton reorganization. The reduced levels of H2S producing enzymes in the PE placentae along with decreased levels of Rho GTPases (Rac1 and Rho A) corroborate the results of PAG and AOAA treatment in down regulating the Rho GTPases in the in vitro grown placental cultures. Reduction of the migratory potential of trophoblastic cells caused due to hypoxia/reoxygenation was rescued by upregulating the H2S expression with the use of NaHS as a H2S donor. DISCUSSION Exogenous H2S increases the migratory potential of the placental cells in culture conditions and also post hypoxia/reoxygenation injury. H2S as a gaso-transmitter holds a great potential as a therapeutic agent. Its long-term effects need to be investigated using model systems (rat/mouse) of PE following it up with clinical regulatory trials.
Collapse
Affiliation(s)
- Sunil Kumar Gupta
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Sankat Mochan
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Pallavi Arora
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Neerja Rani
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Kalpana Luthra
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Sadanand Dwivedi
- Department of Biostatistics, All India Institute of Medical Sciences, New Delhi, India
| | - Neerja Bhatla
- Department of Obstetrics and Gynaecology, All India Institute of Medical Sciences, New Delhi, India
| | - Pallavi Kshetrapal
- Maternal & Child Health, Translational Health Science and Technology Institute, Faridabad, Haryana, India.
| | - Renu Dhingra
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
20
|
Alves P, Amaral C, Teixeira N, Correia-da-Silva G. Effects of a combination of cannabidiol and delta-9-tetrahydrocannabinol on key biological functions of HTR-8/SVneo extravillous trophoblast cells. Toxicology 2023; 495:153614. [PMID: 37567336 DOI: 10.1016/j.tox.2023.153614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/31/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023]
Abstract
In recent years, cannabis use has increased among pregnant women. In addition, the phytocannabinoids cannabidiol (CBD) and delta-9-tetrahydrocannabinol (THC) alone or in combination are being used for therapeutical applications. THC and CBD are able to cross the placenta and a lot remains unknown concerning their impact on angiogenesis and extravillous trophoblasts' (EVTs) migration and invasion, which are essential processes for placentation. Thus, in this study, the HTR-8/SVneo cell line was employed to evaluate the effects of CBD, THC and of their combination (1:1, 2 µM). Cannabinoids affected epithelial-mesenchymal transition, as showed by increased expression of the epithelial protein marker E-cadherin for CBD and CBD plus THC treatments, and decrease of mesenchymal intermediate filament vimentin for all treatments. The gene expression of the metalloproteinases MMP2 and MMP9, and of their inhibitors TIMP1 and TIMP2 was increased, except the latter for THC treatment. Moreover, CBD reduced cell migration and invasion, an effect that was enhanced by its combination with THC. CBD with or without THC also upregulated the gene expression of PGF, while the anti-angiogenic factor sFLT1 was increased for all treatments. VEGFA and FLT1 were not affected. Alone or combined CBD and THC also decreased tube segments' length. Additionally, ERK1/2 and STAT3 phosphorylation was increased in the CBD and CBD plus THC-treated cells, while THC only activated STAT3. AKT activation was only affected by CBD. This work demonstrates that the exposure to cannabinoid-based products containing CBD and/or THC, may interfere with key processes of EVTs differentiation. Therefore, crucial phases of placental development can be affected, compromising pregnancy success.
Collapse
Affiliation(s)
- Patrícia Alves
- UCIBIO.REQUIMTE, Department of Biological Sciences, Laboratory of Biochemistry, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Cristina Amaral
- UCIBIO.REQUIMTE, Department of Biological Sciences, Laboratory of Biochemistry, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Natércia Teixeira
- UCIBIO.REQUIMTE, Department of Biological Sciences, Laboratory of Biochemistry, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Georgina Correia-da-Silva
- UCIBIO.REQUIMTE, Department of Biological Sciences, Laboratory of Biochemistry, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| |
Collapse
|
21
|
Guterstam YC, Acharya G, Schott K, Björkström NK, Gidlöf S, Ivarsson MA. Immune cell profiling of vaginal blood from patients with early pregnancy bleeding. Am J Reprod Immunol 2023; 90:e13738. [PMID: 37491928 DOI: 10.1111/aji.13738] [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: 03/09/2023] [Revised: 05/20/2023] [Accepted: 06/12/2023] [Indexed: 07/27/2023] Open
Abstract
PROBLEM Vaginal bleeding during early pregnancy is estimated to occur in 20% of all pregnancies and it is often difficult to predict who will ultimately miscarry. The role of immune cells in early pregnancy loss is poorly understood. METHOD OF STUDY In this prospective cohort study, 28 pregnant women presenting with first-trimester vaginal bleeding donated vaginal blood, peripheral venous blood, and saliva during their initial emergency room visit, and at a follow-up. The composition, frequency, and phenotype of immune cells in the vaginal blood were determined using flow cytometry. The proteome of serum and saliva was analyzed with OLINK proximity extension assay and correlated to vaginal immune cell phenotype and outcome of pregnancy. The course and outcome of pregnancies were followed and recorded. RESULTS Vaginal blood contained all main immune cell lineages including B cells, NK cells, T cells, and monocytes/macrophages. Notably, vaginal blood immune cells expressed tissue residency markers including CD49a. Women who subsequently miscarried had a higher frequency of vaginal blood CD49a+ NK cells compared to those who did not miscarry, and this correlated with serum levels of granzyme A and H, as well as CSF1, CAIX, and TWEAK. Women in the miscarriage group also had a higher frequency of peripheral blood T cells expressing CD49a. CONCLUSIONS Our study provides novel insight into human reproductive immunology in relation to miscarriage. Tissue-resident NK cells in vaginal blood alone or in combination with serological biomarkers hold potential as prognostic factors in the prediction of pregnancy outcome in women with early pregnancy bleedings.
Collapse
Affiliation(s)
- Ylva Crona Guterstam
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Ganesh Acharya
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Center for Fetal Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Katharina Schott
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Niklas K Björkström
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Sebastian Gidlöf
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Martin A Ivarsson
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
22
|
Yu H, Chen L, Du B. Necroptosis in the pathophysiology of preeclampsia. Cell Cycle 2023; 22:1713-1725. [PMID: 37365800 PMCID: PMC10446795 DOI: 10.1080/15384101.2023.2229138] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 03/30/2023] [Accepted: 06/19/2023] [Indexed: 06/28/2023] Open
Abstract
Necroptosis is a newly-identified form of gene-regulated cell necrosis that is increasingly considered to be a pathway associated with human pathophysiological conditions. Cells undergoing necroptosis exhibit necrotic phenotypes, including disruption of the plasma membrane integrity, organelle swelling, and cytolysis. Accumulating evidence suggests that trophoblast necroptosis plays a complex role in preeclampsia (PE). However, the exact pathogenesis remains unclear. Its unique mechanisms of action in various diseases are expected to provide prospects for the treatment of PE. Therefore, it is necessary to further explore its molecular mechanism in PE in order to identify potential therapeutic options. This review examines the current knowledge regarding the role and mechanisms of necroptosis in PE and provides a theoretical basis for new therapeutic targets for PE.
Collapse
Affiliation(s)
- Hongbiao Yu
- Department of Obstetrics and Gynecology, the Second Clinical College of North Sichuan Medical College, Nanchong Central Hospital, Nanchong, Sichuan, China
| | - Ling Chen
- Department of Oncology, the Second Clinical College of North Sichuan Medical College, Nanchong Central Hospital, Nanchong, Sichuan, China
| | - Boyu Du
- Department of Obstetrics and Gynecology, the Second Clinical College of North Sichuan Medical College, Nanchong Central Hospital, Nanchong, Sichuan, China
| |
Collapse
|
23
|
Tousty P, Fraszczyk-Tousty M, Dzidek S, Jasiak-Jóźwik H, Michalczyk K, Kwiatkowska E, Cymbaluk-Płoska A, Torbé A, Kwiatkowski S. Low-Dose Aspirin after ASPRE-More Questions Than Answers? Current International Approach after PE Screening in the First Trimester. Biomedicines 2023; 11:1495. [PMID: 37371598 DOI: 10.3390/biomedicines11061495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/16/2023] [Accepted: 05/18/2023] [Indexed: 06/29/2023] Open
Abstract
Preeclampsia (PE) is a multi-factorial disorder of pregnancy, and it continues to be one of the leading causes of fetal and maternal morbidity and mortality worldwide. Aspirin is universally recommended for high-risk women to reduce preeclampsia risk. The purpose of this review is to summarize the recommendations of various scientific societies on predicting preeclampsia and their indications for the inclusion of acetylsalicylic acid (ASA) prophylaxis. Fourteen guidelines were compared. The recommended dose, screening method, and gestational age at the start of the test vary depending on the recommendation. The societies are inclined to recommend using increasingly higher doses (>75 mg) of ASA, with many encouraging doses from 100 mg upward. Most societies indicate that the optimal time for implementing aspirin is prior to 16 weeks' gestation. Following the publication of the Aspirin for Evidence-Based Preeclampsia Prevention (ASPRE) trial results and other papers evaluating the Fetal Medicine Foundation (FMF) screening model, a large number of societies have changed their recommendations from those based on risk factors alone to the ones based on the risk assessment proposed by the FMF. This allows for the detection of a high-risk pregnancy population in whom aspirin will be remarkably effective in preventing preterm PE, thereby decreasing maternal and fetal morbidity.
Collapse
Affiliation(s)
- Piotr Tousty
- Department of Gynecology and Obstetrics, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Magda Fraszczyk-Tousty
- Department of Neonatology and Neonatal Intensive Care, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Sylwia Dzidek
- Department of Gynecology and Obstetrics, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Hanna Jasiak-Jóźwik
- Department of Gynecology and Obstetrics, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Kaja Michalczyk
- Department of Gynecological Surgery and Gynecological Oncology of Adults and Adolescents, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Ewa Kwiatkowska
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Aneta Cymbaluk-Płoska
- Department of Reconstructive Surgery and Gynecological Oncology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Andrzej Torbé
- Department of Gynecology and Obstetrics, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Sebastian Kwiatkowski
- Department of Gynecology and Obstetrics, Pomeranian Medical University, 70-111 Szczecin, Poland
| |
Collapse
|
24
|
Huang CC, Hsueh YW, Chang CW, Hsu HC, Yang TC, Lin WC, Chang HM. Establishment of the fetal-maternal interface: developmental events in human implantation and placentation. Front Cell Dev Biol 2023; 11:1200330. [PMID: 37266451 PMCID: PMC10230101 DOI: 10.3389/fcell.2023.1200330] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/09/2023] [Indexed: 06/03/2023] Open
Abstract
Early pregnancy is a complex and well-orchestrated differentiation process that involves all the cellular elements of the fetal-maternal interface. Aberrant trophoblast-decidual interactions can lead to miscarriage and disorders that occur later in pregnancy, including preeclampsia, intrauterine fetal growth restriction, and preterm labor. A great deal of research on the regulation of implantation and placentation has been performed in a wide range of species. However, there is significant species variation regarding trophoblast differentiation as well as decidual-specific gene expression and regulation. Most of the relevant information has been obtained from studies using mouse models. A comprehensive understanding of the physiology and pathology of human implantation and placentation has only recently been obtained because of emerging advanced technologies. With the derivation of human trophoblast stem cells, 3D-organoid cultures, and single-cell analyses of differentiated cells, cell type-specific transcript profiles and functions were generated, and each exhibited a unique signature. Additionally, through integrative transcriptomic information, researchers can uncover the cellular dysfunction of embryonic and placental cells in peri-implantation embryos and the early pathological placenta. In fact, the clinical utility of fetal-maternal cellular trafficking has been applied for the noninvasive prenatal diagnosis of aneuploidies and the prediction of pregnancy complications. Furthermore, recent studies have proposed a viable path toward the development of therapeutic strategies targeting placenta-enriched molecules for placental dysfunction and diseases.
Collapse
|
25
|
Biyik I, Metineren H, Ozturk E, Simsek S, Oztas E, Guldur ME, Ince O. Placental T-Cadherin Correlates With Trophoblastic Invasion Anomalies: Placenta Percreta and Fetal Growth Restriction. Int J Gynecol Pathol 2023; 42:293-300. [PMID: 36731049 DOI: 10.1097/pgp.0000000000000884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In this study, we compared the placental T-cadherin staining intensity of pregnant women with placenta percreta (PP) and asymmetrical fetal growth restriction (FGR) compared with healthy control pregnancies. Placental T-cadherin levels of the placenta of 86 pregnant women in total, 25 with FGR, 30 with healthy pregnant subjects, and 31 with PP, were examined using monoclonal anti-T-cadherin (CDH13) antibody for immunohistochemical examination. In immunohistochemistry, H -scores were used for each group to compare the expression of T-cadherin in extravillous trophoblast (EVT) cells. T-cadherin H -score of EVTs was highest in the FGR group and the lowest in the PP group. The difference in H -score between the FGR group and the control group was not statistically significant ( P =0.344). The difference between the PP group and the other 2 groups was significant ( P <0.0001). Multivariable linear regression analysis with a stepwise elimination method was performed in order to identify demographic and clinical parameters with significant effects on the T-cadherin H -score of EVTs. The estimation results identified only the disease group as a significant predictor of the H -score of EVTs ( R2 =0.340, P <0.0001). The highest T-cadherin H -score of EVTs was found in the FGR group and the lowest in the PP group. The low T-cadherin H-score values in the PP group suggest that low T-cadherin EVTs may be associated with increased placental invasion. Likewise, despite the statistical insignificance, a higher T-cadherin H -score of EVTs in FGR compared with controls implies a decreased invasiveness of the placenta in FGR.
Collapse
|
26
|
Arutyunyan A, Roberts K, Troulé K, Wong FCK, Sheridan MA, Kats I, Garcia-Alonso L, Velten B, Hoo R, Ruiz-Morales ER, Sancho-Serra C, Shilts J, Handfield LF, Marconato L, Tuck E, Gardner L, Mazzeo CI, Li Q, Kelava I, Wright GJ, Prigmore E, Teichmann SA, Bayraktar OA, Moffett A, Stegle O, Turco MY, Vento-Tormo R. Spatial multiomics map of trophoblast development in early pregnancy. Nature 2023; 616:143-151. [PMID: 36991123 PMCID: PMC10076224 DOI: 10.1038/s41586-023-05869-0] [Citation(s) in RCA: 125] [Impact Index Per Article: 62.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 02/21/2023] [Indexed: 03/31/2023]
Abstract
The relationship between the human placenta-the extraembryonic organ made by the fetus, and the decidua-the mucosal layer of the uterus, is essential to nurture and protect the fetus during pregnancy. Extravillous trophoblast cells (EVTs) derived from placental villi infiltrate the decidua, transforming the maternal arteries into high-conductance vessels1. Defects in trophoblast invasion and arterial transformation established during early pregnancy underlie common pregnancy disorders such as pre-eclampsia2. Here we have generated a spatially resolved multiomics single-cell atlas of the entire human maternal-fetal interface including the myometrium, which enables us to resolve the full trajectory of trophoblast differentiation. We have used this cellular map to infer the possible transcription factors mediating EVT invasion and show that they are preserved in in vitro models of EVT differentiation from primary trophoblast organoids3,4 and trophoblast stem cells5. We define the transcriptomes of the final cell states of trophoblast invasion: placental bed giant cells (fused multinucleated EVTs) and endovascular EVTs (which form plugs inside the maternal arteries). We predict the cell-cell communication events contributing to trophoblast invasion and placental bed giant cell formation, and model the dual role of interstitial EVTs and endovascular EVTs in mediating arterial transformation during early pregnancy. Together, our data provide a comprehensive analysis of postimplantation trophoblast differentiation that can be used to inform the design of experimental models of the human placenta in early pregnancy.
Collapse
Affiliation(s)
- Anna Arutyunyan
- Wellcome Sanger Institute, Cambridge, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | | | | | | | - Megan A Sheridan
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Ilia Kats
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Britta Velten
- Wellcome Sanger Institute, Cambridge, UK
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Regina Hoo
- Wellcome Sanger Institute, Cambridge, UK
| | | | | | | | | | - Luca Marconato
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | | | - Lucy Gardner
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | | | - Qian Li
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Iva Kelava
- Wellcome Sanger Institute, Cambridge, UK
| | - Gavin J Wright
- Department of Biology, Hull York Medical School, York Biomedical Research Institute, University of York, York, UK
| | | | - Sarah A Teichmann
- Wellcome Sanger Institute, Cambridge, UK
- Theory of Condensed Matter, Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, UK
| | | | - Ashley Moffett
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
- Department of Pathology, University of Cambridge, Cambridge, UK.
| | - Oliver Stegle
- Wellcome Sanger Institute, Cambridge, UK.
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany.
| | - Margherita Y Turco
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
- Department of Pathology, University of Cambridge, Cambridge, UK.
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland.
| | - Roser Vento-Tormo
- Wellcome Sanger Institute, Cambridge, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| |
Collapse
|
27
|
Abstract
Pre-eclampsia is a life-threatening disease of pregnancy unique to humans and a leading cause of maternal and neonatal morbidity and mortality. Women who survive pre-eclampsia have reduced life expectancy, with increased risks of stroke, cardiovascular disease and diabetes, while babies from a pre-eclamptic pregnancy have increased risks of preterm birth, perinatal death and neurodevelopmental disability and cardiovascular and metabolic disease later in life. Pre-eclampsia is a complex multisystem disease, diagnosed by sudden-onset hypertension (>20 weeks of gestation) and at least one other associated complication, including proteinuria, maternal organ dysfunction or uteroplacental dysfunction. Pre-eclampsia is found only when a placenta is or was recently present and is classified as preterm (delivery <37 weeks of gestation), term (delivery ≥37 weeks of gestation) and postpartum pre-eclampsia. The maternal syndrome of pre-eclampsia is driven by a dysfunctional placenta, which releases factors into maternal blood causing systemic inflammation and widespread maternal endothelial dysfunction. Available treatments target maternal hypertension and seizures, but the only 'cure' for pre-eclampsia is delivery of the dysfunctional placenta and baby, often prematurely. Despite decades of research, the aetiology of pre-eclampsia, particularly of term and postpartum pre-eclampsia, remains poorly defined. Significant advances have been made in the prediction and prevention of preterm pre-eclampsia, which is predicted in early pregnancy through combined screening and is prevented with daily low-dose aspirin, starting before 16 weeks of gestation. By contrast, the prediction of term and postpartum pre-eclampsia is limited and there are no preventive treatments. Future research must investigate the pathogenesis of pre-eclampsia, in particular of term and postpartum pre-eclampsia, and evaluate new prognostic tests and treatments in adequately powered clinical trials.
Collapse
|
28
|
Dall'Asta A, Melito C, Morganelli G, Lees C, Ghi T. Determinants of placental insufficiency in fetal growth restriction. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2023; 61:152-157. [PMID: 36349884 DOI: 10.1002/uog.26111] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/14/2022] [Accepted: 10/18/2022] [Indexed: 05/27/2023]
Affiliation(s)
- A Dall'Asta
- Department of Medicine and Surgery, Obstetrics and Gynaecology Unit, University of Parma, Parma, Italy
- Department of Metabolism, Digestion and Reproduction, Institute of Reproductive and Developmental Biology, Imperial College London, London, UK
| | - C Melito
- Department of Medicine and Surgery, Obstetrics and Gynaecology Unit, University of Parma, Parma, Italy
- Department of Obstetrics and Gynaecology, IRCCS Fondazione Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - G Morganelli
- Department of Medicine and Surgery, Obstetrics and Gynaecology Unit, University of Parma, Parma, Italy
| | - C Lees
- Department of Metabolism, Digestion and Reproduction, Institute of Reproductive and Developmental Biology, Imperial College London, London, UK
- Centre for Fetal Care, Department of Obstetrics and Gynaecology, Queen Charlotte's and Chelsea Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - T Ghi
- Department of Medicine and Surgery, Obstetrics and Gynaecology Unit, University of Parma, Parma, Italy
| |
Collapse
|
29
|
Siriwardena D, Boroviak TE. Evolutionary divergence of embryo implantation in primates. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210256. [PMID: 36252209 DOI: 10.1098/rstb.2021.0256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Implantation of the conceptus into the uterus is absolutely essential for successful embryo development. In humans, our understanding of this process has remained rudimentary owing to the inaccessibility of early implantation stages. Non-human primates recapitulate many aspects of human embryo development and provide crucial insights into trophoblast development, uterine receptivity and embryo invasion. Moreover, primate species exhibit a variety of implantation strategies and differ in embryo invasion depths. This review examines conservation and divergence of the key processes required for embryo implantation in different primates and in comparison with the canonical rodent model. We discuss trophectoderm compartmentalization, endometrial remodelling and embryo adhesion and invasion. Finally, we propose that studying the mechanism controlling invasion depth between different primate species may provide new insights and treatment strategies for placentation disorders in humans. This article is part of the theme issue 'Extraembryonic tissues: exploring concepts, definitions and functions across the animal kingdom'.
Collapse
Affiliation(s)
- 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
| | - 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
|
30
|
Reliability of Rodent and Rabbit Models in Preeclampsia Research. Int J Mol Sci 2022; 23:ijms232214344. [PMID: 36430816 PMCID: PMC9696504 DOI: 10.3390/ijms232214344] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/14/2022] [Accepted: 11/14/2022] [Indexed: 11/22/2022] Open
Abstract
In vivo studies on the pathology of gestation, including preeclampsia, often use small mammals such as rabbits or rodents, i.e., mice, rats, hamsters, and guinea pigs. The key advantage of these animals is their short reproductive cycle; in addition, similar to humans, they also develop a haemochorial placenta and present a similar transformation of maternal spiral arteries. Interestingly, pregnant dams also demonstrate a similar reaction to inflammatory factors and placentally derived antiangiogenic factors, i.e., soluble fms-like tyrosine kinase 1 (sFlt-1) or soluble endoglin-1 (sEng), as preeclamptic women: all animals present an increase in blood pressure and usually proteinuria. These constitute the classical duet that allows for the recognition of preeclampsia. However, the time of initiation of maternal vessel remodelling and the depth of trophoblast invasion differs between rabbits, rodents, and humans. Unfortunately, at present, no known animal replicates a human pregnancy exactly, and hence, the use of rabbit and rodent models is restricted to the investigation of individual aspects of human gestation only. This article compares the process of placentation in rodents, rabbits, and humans, which should be considered when planning experiments on preeclampsia; these aspects might determine the success, or failure, of the study. The report also reviews the rodent and rabbit models used to investigate certain aspects of the pathomechanism of human preeclampsia, especially those related to incorrect trophoblast invasion, placental hypoxia, inflammation, or maternal endothelial dysfunction.
Collapse
|
31
|
Brosens JJ, Bennett PR, Abrahams VM, Ramhorst R, Coomarasamy A, Quenby S, Lucas ES, McCoy RC. Maternal selection of human embryos in early gestation: Insights from recurrent miscarriage. Semin Cell Dev Biol 2022; 131:14-24. [PMID: 35094946 PMCID: PMC9325922 DOI: 10.1016/j.semcdb.2022.01.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/21/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023]
Abstract
Compared to most mammals, human pregnancy is unusual in that it involves chromosomally diverse embryos, cyclical breakdown and regeneration of the uterine mucosa, and intimate integration of fetal and maternal cells at the uteroplacental interface. Not surprisingly, pregnancy often falters in early gestation. Whether these losses result in clinical miscarriages depends on the origins and impacts of chromosomal errors on fetal development and the ability of the decidualizing endometrium to engage in embryo biosensing and selection. Aneuploidy originating in oocytes during meiosis drives the age-related risk of miscarriage. By contrast, the frequency of endometrial cycles with an impaired decidual response may account for the stepwise increase in miscarriage rates with each pregnancy loss independently of maternal age. Additional physiological mechanisms operate in early gestation to ensure that most failing pregnancies are lost before vascular maternal-fetal connections are established by the end of the first trimester. Here, we summarise how investigations into the mechanisms that cause miscarriage led to new insights into the processes that govern maternal selection of human embryos in early gestation.
Collapse
Affiliation(s)
- Jan J Brosens
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK; Tommy's National Centre for Miscarriage Research, University Hospitals Coventry & Warwickshire NHS Trust, Coventry CV2 2DX, UK.
| | - Phillip R Bennett
- Tommy's National Centre for Miscarriage Research, Imperial College London, UK
| | - Vikki M Abrahams
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University, New Haven, CT, USA
| | - Rosanna Ramhorst
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales IQUIBICEN, Buenos Aires, Argentina
| | - Arri Coomarasamy
- Tommy's National Centre for Miscarriage Research, Institute of Metabolism and Systems Research, University of Birmingham, UK
| | - Siobhan Quenby
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK; Tommy's National Centre for Miscarriage Research, University Hospitals Coventry & Warwickshire NHS Trust, Coventry CV2 2DX, UK
| | - Emma S Lucas
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, UK
| | - Rajiv C McCoy
- Department of Biology, Johns Hopkins University, Baltimore, Maryland 21218, USA
| |
Collapse
|
32
|
Cindrova-Davies T, Sferruzzi-Perri AN. Human placental development and function. Semin Cell Dev Biol 2022; 131:66-77. [PMID: 35393235 DOI: 10.1016/j.semcdb.2022.03.039] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/30/2022] [Accepted: 03/30/2022] [Indexed: 12/14/2022]
Abstract
The placenta is a transient fetal organ that plays a critical role in the health and wellbeing of both the fetus and its mother. Functionally, the placenta sustains the growth of the fetus as it facilitates delivery of oxygen and nutrients and removal of waste products. Not surprisingly, defective early placental development is the primary cause of common disorders of pregnancy, including recurrent miscarriage, fetal growth restriction, pre-eclampsia and stillbirth. Adverse pregnancy conditions will also affect the life-long health of the fetus via developmental programming[1]. Despite its critical importance in reproductive success and life-long health, our understanding of placental development is not extensive, largely due to ethical limitations to studying early or chronological placental development, lack of long-term in vitro models, or comparative animal models. In this review, we examine current knowledge of early human placental development, discuss the critical role of the maternal endometrium and of the fetal-maternal dialogue in pregnancy success, and we explore the latest models of trophoblast and endometrial stem cells. In addition, we discuss the role of oxygen in placental formation and function, how nutrient delivery is mediated during the periods of histotrophic nutrition (uptake of uterine secretions) and haemotrophic nutrition (exchange between the maternal and fetal circulations), and how placental endocrine function facilitates fetal growth and development.
Collapse
Affiliation(s)
- Tereza Cindrova-Davies
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Amanda N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK.
| |
Collapse
|
33
|
Zhang B, Zhang F, Lu F, Wang J, Zhou W, Wang H, Yu B. Reduced cell invasion may be a characteristic of placental defects in pregnant women of advanced maternal age at single-cell level. J Zhejiang Univ Sci B 2022; 23:747-759. [PMID: 36111571 DOI: 10.1631/jzus.b2101024] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The mechanisms underlying pregnancy complications caused by advanced maternal age (AMA) remain unclear. We analyzed the cellular signature and transcriptomes of human placentas in AMA women to elucidate these mechanisms. Placental tissues from two AMA women and two controls were used for single-cell RNA-sequencing (scRNA-seq). Controls consisted of AMA women who did not experience any pregnancy complications and pregnant women below the age of 35 years without pregnancy complications. Trophoblast cells were obtained from the placentas of another six pregnant women (three AMA women and three controls), and in-vitro transwell assays were conducted to observe the cell invasion ability. Thirty additional samples (from 15 AMA women and 15 controls) were analyzed to verify the specific expression of serine protease inhibitor clade E member 1 (SERPINE1). Preliminary study of the role of SERPINE1 in cell invasion was carried out with HTR8-S/Vneo cells. High-quality transcriptomes of 27 607 cells were detected. Three types of trophoblast cells were detected, which were further classified into eight subtypes according to differences in gene expression and Gene Ontology (GO) function. We identified 110 differentially expressed genes (DEGs) in trophoblast cells between the AMA and control groups, and the DEGs were enriched in multiple pathways related to cell invasion. In-vitro transwell assays suggested that the invading trophoblast cells in AMA women were reduced. SERPINE1 was specifically expressed in the trophoblast, and its expression was higher in AMA women (P<0.05). Transfection of human SERPINE1 (hSERPINE1) into HTR8-S/Vneo trophoblast cells showed fewer invading cells in the hSERPINE1 group. Impaired cell invasion may underlie the increased risk of adverse pregnancy outcomes in AMA women. Abnormal expression of SERPINE1 in extravillous trophoblast (EVT) cells appears to play an important role.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Medical Genetics, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Feng Zhang
- Department of Medical Genetics, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Fengying Lu
- Department of Medical Genetics, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Jing Wang
- Department of Medical Genetics, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Wenbai Zhou
- Department of Medical Genetics, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Huihui Wang
- Department of Medical Genetics, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China
| | - Bin Yu
- Department of Medical Genetics, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213000, China.
| |
Collapse
|
34
|
de Vos ES, Koning AHJ, Steegers-Theunissen RPM, Willemsen SP, van Rijn BB, Steegers EAP, Mulders AGMGJ. Assessment of first-trimester utero-placental vascular morphology by 3D power Doppler ultrasound image analysis using a skeletonization algorithm: the Rotterdam Periconception Cohort. Hum Reprod 2022; 37:2532-2545. [PMID: 36125007 PMCID: PMC9627684 DOI: 10.1093/humrep/deac202] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 08/16/2022] [Indexed: 11/22/2022] Open
Abstract
STUDY QUESTION Can three-dimensional (3D) Power Doppler (PD) ultrasound and a skeletonization algorithm be used to assess first-trimester development of the utero-placental vascular morphology? SUMMARY ANSWER The application of 3D PD ultrasonography and a skeletonization algorithm facilitates morphologic assessment of utero-placental vascular development in the first trimester and reveals less advanced vascular morphologic development in pregnancies with placenta-related complications than in pregnancies without placenta-related complications. WHAT IS KNOWN ALREADY Suboptimal development of the utero-placental vasculature is one of the main contributors to the periconceptional origin of placenta-related complications. The nature and attribution of aberrant vascular structure and branching patterns remain unclear, as validated markers monitoring first-trimester utero-placental vascular morphologic development are lacking. STUDY DESIGN, SIZE, DURATION In this prospective observational cohort, 214 ongoing pregnancies were included before 10 weeks gestational age (GA) at a tertiary hospital between January 2017 and July 2018, as a subcohort of the ongoing Rotterdam Periconception Cohort study. PARTICIPANTS/MATERIALS, SETTING, METHODS By combining 3D PD ultrasonography and virtual reality, utero-placental vascular volume (uPVV) measurements were obtained at 7, 9 and 11 weeks GA. A skeletonization algorithm was applied to the uPVV measurements to generate the utero-placental vascular skeleton (uPVS), a network-like structure containing morphologic characteristics of the vasculature. Quantification of vascular morphology was performed by assigning a morphologic characteristic to each voxel in the uPVS (end-, vessel-, bifurcation- or crossing-point) and calculating total vascular network length. A Mann–Whitney U test was performed to investigate differences in morphologic development of the first-trimester utero-placental vasculature between pregnancies with and without placenta-related complications. Linear mixed models were used to estimate trajectories of the morphologic characteristics in the first trimester. MAIN RESULTS AND THE ROLE OF CHANCE All morphologic characteristics of the utero-placental vasculature increased significantly in the first trimester (P < 0.005). In pregnancies with placenta-related complications (n = 54), utero-placental vascular branching was significantly less advanced at 9 weeks GA (vessel points P = 0.040, bifurcation points P = 0.050, crossing points P = 0.020, total network length P = 0.023). Morphologic growth trajectories remained similar after adjustment for parity, conception mode, foetal sex and occurrence of placenta-related complications. LIMITATIONS, REASONS FOR CAUTION The tertiary setting of this prospective observational study provides high internal, but possibly limited external, validity. Extrapolation of the study’s findings should therefore be addressed with caution. WIDER IMPLICATIONS OF THE FINDINGS The uPVS enables assessment of morphologic development of the first-trimester utero-placental vasculature. Further investigation of this innovative methodology needs to determine its added value for the assessment of (patho-) physiological utero-placental vascular development. STUDY FUNDING/COMPETING INTEREST(S) This research was funded by the Department of Obstetrics and Gynecology of the Erasmus MC, University Medical Centre, Rotterdam, The Netherlands. There are no conflicts of interest. TRIAL REGISTRATION NUMBER Registered at the Dutch Trial Register (NTR6854).
Collapse
Affiliation(s)
- Eline S de Vos
- Department of Obstetrics and Gynecology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Anton H J Koning
- Department of Pathology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | | | - Sten P Willemsen
- Department of Obstetrics and Gynecology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands.,Department of Biostatistics, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Bas B van Rijn
- Department of Obstetrics and Gynecology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Eric A P Steegers
- Department of Obstetrics and Gynecology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Annemarie G M G J Mulders
- Department of Obstetrics and Gynecology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
35
|
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: 22] [Impact Index Per Article: 7.3] [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
|
36
|
Nikitina TV, Lebedev IN. Stem Cell-Based Trophoblast Models to Unravel the Genetic Causes of Human Miscarriages. Cells 2022; 11:1923. [PMID: 35741051 PMCID: PMC9221414 DOI: 10.3390/cells11121923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/10/2022] [Accepted: 06/12/2022] [Indexed: 02/01/2023] Open
Abstract
Miscarriage affects approximately 15% of clinically recognized pregnancies, and 1-3% of couples experience pregnancy loss recurrently. Approximately 50-60% of miscarriages result from chromosomal abnormalities, whereas up to 60% of euploid recurrent abortions harbor variants in candidate genes. The growing number of detected genetic variants requires an investigation into their role in adverse pregnancy outcomes. Since placental defects are the main cause of first-trimester miscarriages, the purpose of this review is to provide a survey of state-of-the-art human in vitro trophoblast models that can be used for the functional assessment of specific abnormalities/variants implicated in pregnancy loss. Since 2018, when primary human trophoblast stem cells were first derived, there has been rapid growth in models of trophoblast lineage. It has been found that a proper balance between self-renewal and differentiation in trophoblast progenitors is crucial for the maintenance of pregnancy. Different responses to aneuploidy have been shown in human embryonic and extra-embryonic lineages. Stem cell-based models provide a powerful tool to explore the effect of a specific aneuploidy/variant on the fetus through placental development, which is important, from a clinical point of view, for deciding on the suitability of embryos for transfer after preimplantation genetic testing for aneuploidy.
Collapse
Affiliation(s)
- Tatiana V. Nikitina
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, 634050 Tomsk, Russia;
| | | |
Collapse
|
37
|
Reactive Oxygen Species are Essential for Placental Angiogenesis During Early Gestation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4290922. [PMID: 35693704 PMCID: PMC9177322 DOI: 10.1155/2022/4290922] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 05/07/2022] [Indexed: 12/30/2022]
Abstract
Background Preeclampsia (PE) is associated with insufficient placental perfusion attributed to maldevelopment of the placental vasculature. Reactive oxygen species (ROS) are implicated in angiogenesis, but their regulatory effects and mechanisms in placental vascular development remain unclear. Methods Placental oxidative stress was determined throughout gestation by measuring 4-hydroxynonenal (4HNE) and malondialdehyde (MDA). The antioxidant MitoQ was administered to pregnant mice from GDs 7.5 to 11.5; placental morphology and angiogenesis pathways were examined on GDs 11.5 and 18.5. Moreover, we established a mouse mFlt-1-induced PE model and assessed blood pressure, urine protein levels, and placental vascular development on GDs 11.5 and 18.5. Human umbilical vein endothelial cells (HUVECs) were treated with various H2O2 concentrations to evaluate cell viability, intracellular ROS levels, and tube formation capability. MitoQ, an AKT inhibitor and an ERK1/2 inhibitor were applied to validate the ROS-mediated mechanism regulating placental angiogenesis. Results First-trimester placentas presented significantly higher MDA and 4HNE levels. MitoQ significantly reduced the blood vessel density and angiogenesis pathway activity in the placenta on GDs 11.5 and 18.5. Serum sFlt-1 levels were elevated, and we observed poor placental angiogenesis and PE-like symptoms in cases with mFlt-1 overexpression. Moderate H2O2 treatment promoted HUVEC proliferation and angiogenesis, whereas these improvements were abolished by MitoQ, AKT inhibitor, or ERK1/2 inhibitor treatment. Conclusions Moderate ROS levels are essential for placental angiogenesis; diminishing ROS with potent antioxidants during placentation decreases placental angiogenesis and increases PE risk. Therefore, antioxidant therapy should be considered carefully for normal pregnant women during early gestation.
Collapse
|
38
|
Petroff MG, Nguyen SL, Ahn SH. Fetal‐placental
antigens and the maternal immune system: Reproductive immunology comes of age. Immunol Rev 2022; 308:25-39. [PMID: 35643905 PMCID: PMC9328203 DOI: 10.1111/imr.13090] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 05/07/2022] [Accepted: 05/11/2022] [Indexed: 12/20/2022]
Abstract
Reproductive physiology and immunology as scientific disciplines each have rich, largely independent histories. The physicians and philosophers of ancient Greece made remarkable observations and inferences to explain regeneration as well as illness and immunity. The scientific enlightenment of the renaissance and the technological advances of the past century have led to the explosion of knowledge that we are experiencing today. Breakthroughs in transplantation, immunology, and reproduction eventually culminated with Medawar’s discovery of acquired immunological tolerance, which helped to explain the transplantation success and failure. Medawar’s musings also keenly pointed out that the fetus apparently breaks these newly discovered rules, and with this, the field of reproductive immunology was launched. As a result of having stemmed from transplantation immunology, scientist still analogizes the fetus to a successful allograft. Although we now know of the fundamental differences between the two, this analogy remains a useful tool to understand how the fetus thrives despite its immunological disparity with the mother. Here, we review the history of reproductive immunology, and how major and minor histocompatibility antigens, blood group antigens, and tissue‐specific “self” antigens from the fetus and transplanted organs parallel and differ.
Collapse
Affiliation(s)
- Margaret G. Petroff
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine Michigan State University East Lansing Michigan USA
- Departments of Microbiology and Molecular Genetics, College of Veterinary Medicine and College of Human Medicine Michigan State University East Lansing Michigan USA
- Cell and Molecular Biology Program, College of Natural Science Michigan State University East Lansing Michigan USA
| | - Sean L. Nguyen
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine Michigan State University East Lansing Michigan USA
- Cell and Molecular Biology Program, College of Natural Science Michigan State University East Lansing Michigan USA
| | - Soo Hyun Ahn
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine Michigan State University East Lansing Michigan USA
| |
Collapse
|
39
|
Jauniaux E, Jurkovic D, Hussein AM, Burton GJ. New insights into the etiopathology of placenta accreta spectrum. Am J Obstet Gynecol 2022; 227:384-391. [PMID: 35248577 DOI: 10.1016/j.ajog.2022.02.038] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/07/2022] [Accepted: 02/21/2022] [Indexed: 11/19/2022]
Abstract
Placenta accreta has been described as a spectrum of abnormal attachment of villous tissue to the uterine wall, ranging from superficial attachment to the inner myometrium without interposing decidua to transmural invasion through the entire uterine wall and beyond. These descriptions have prevailed for more than 50 years and form the basis for the diagnosis and grading of accreta placentation. Accreta placentation is essentially the consequence of uterine remodeling after surgery, primarily after cesarean delivery. Large cesarean scar defects in the lower uterine segment are associated with failure of normal decidualization and loss of the subdecidual myometrium. These changes allow the placental anchoring villi to implant, and extravillous trophoblast cells to migrate, close to the serosal surface of the uterus. These microscopic features are central to the misconception that the accreta placental villous tissue is excessively invasive and have led to much confusion and heterogeneity in clinical data. Progressive recruitment of large arteries in the uterine wall, that is, helicine, arcuate, and/or radial arteries, results in high-velocity maternal blood entering the intervillous space from the first trimester of pregnancy and subsequent formation of placental lacunae. Recently, guided sampling of accreta areas at delivery has enabled accurate correlation of prenatal imaging data with intraoperative features and histopathologic findings. In more than 70% of samples, there were thick fibrinoid depositions between the tip of most anchoring villi and the underlying uterine wall and around all deeply implanted villi. The distortion of the uteroplacental interface by these dense depositions and the loss of the normal plane of separation are the main factors leading to abnormal placental attachment. These data challenged the classical concept that placenta accreta is simply owing to villous tissue sitting atop the superficial myometrium without interposed decidua. Moreover, there is no evidence in accreta placentation that the extravillous trophoblast is abnormally invasive or that villous tissue can cross the uterine serosa into the pelvis. It is the size of the scar defect, the amount of placental tissue developing inside the scar, and the residual myometrial thickness in the scar area that determine the distance between the placental basal plate and the uterine serosa and thus the risk of accreta placentation.
Collapse
Affiliation(s)
- Eric Jauniaux
- Faculty of Population Health Sciences, Elizabeth Garrett Anderson Institute for Women's Health, Faculty of Population Health Sciences, London, United Kingdom.
| | - Davor Jurkovic
- Faculty of Population Health Sciences, Elizabeth Garrett Anderson Institute for Women's Health, Faculty of Population Health Sciences, London, United Kingdom
| | - Ahmed M Hussein
- Department of Obstetrics and Gynecology, University of Cairo, Cairo, Egypt
| | - Graham J Burton
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
40
|
Redman CW, Staff AC, Roberts JM. Syncytiotrophoblast stress in preeclampsia: the convergence point for multiple pathways. Am J Obstet Gynecol 2022; 226:S907-S927. [PMID: 33546842 DOI: 10.1016/j.ajog.2020.09.047] [Citation(s) in RCA: 185] [Impact Index Per Article: 61.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/14/2020] [Accepted: 09/19/2020] [Indexed: 12/29/2022]
Abstract
Preeclampsia evolves in 2 stages: a placental problem that generates signals to the mother to cause a range of responses that comprise the second stage (preeclampsia syndrome). The first stage of early-onset preeclampsia is poor placentation, which we here call malplacentation. The spiral arteries are incompletely remodeled, leading to later placental malperfusion, relatively early in the second half of pregnancy. The long duration of the first stage (several months) is unsurprisingly associated with fetal growth restriction. The first stage of late-onset preeclampsia, approximately 80% of total cases, is shorter (several weeks) and part of a process that is common to all pregnancies. Placental function declines as it outgrows uterine capacity, with increasing chorionic villous packing, compression of the intervillous space, and fetal hypoxia, and causes late-onset clinical presentations such as "unexplained" stillbirths, late-onset fetal growth restriction, or preeclampsia. The second stages of early- and late-onset preeclampsia share syncytiotrophoblast stress as the most relevant feature that causes the maternal syndrome. Syncytiotrophoblast stress signals in the maternal circulation are probably the most specific biomarkers for preeclampsia. In addition, soluble fms-like tyrosine kinase-1 (mainly produced by syncytiotrophoblast) is the best-known biomarker and is routinely used in clinical practice in many locations. How the stress signals change over time in normal pregnancies indicates that syncytiotrophoblast stress begins on average at 30 to 32 weeks' gestation and progresses to term. At term, syncytiotrophoblast shows increasing markers of stress, including apoptosis, pyroptosis, autophagy, syncytial knots, and necrosis. We label this phenotype the "twilight placenta" and argue that it accounts for the clinical problems of postmature pregnancies. Senescence as a stress response differs in multinuclear syncytiotrophoblast from that of mononuclear cells. Syncytiotrophoblast irreversibly acquires part of the senescence phenotype (cell cycle arrest) when it is formed by cell fusion. The 2 pathways converge on the common pathologic endpoint, syncytiotrophoblast stress, and contribute to preeclampsia subtypes. We highlight that the well-known heterogeneity of the preeclampsia syndrome arises from different pathways to this common endpoint, influenced by maternal genetics, epigenetics, lifestyle, and environmental factors with different fetal and maternal responses to the ensuing insults. This complexity mandates a reassessment of our approach to predicting and preventing preeclampsia, and we summarize research priorities to maximize what we can learn about these important issues.
Collapse
|
41
|
Aye IL, Aiken CE, Charnock-Jones DS, Smith GC. Placental energy metabolism in health and disease-significance of development and implications for preeclampsia. Am J Obstet Gynecol 2022; 226:S928-S944. [PMID: 33189710 DOI: 10.1016/j.ajog.2020.11.005] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 02/08/2023]
Abstract
The placenta is a highly metabolically active organ fulfilling the bioenergetic and biosynthetic needs to support its own rapid growth and that of the fetus. Placental metabolic dysfunction is a common occurrence in preeclampsia although its causal relationship to the pathophysiology is unclear. At the outset, this may simply be seen as an "engine out of fuel." However, placental metabolism plays a vital role beyond energy production and is linked to physiological and developmental processes. In this review, we discuss the metabolic basis for placental dysfunction and propose that the alterations in energy metabolism may explain many of the placental phenotypes of preeclampsia such as reduced placental and fetal growth, redox imbalance, oxidative stress, altered epigenetic and gene expression profiles, and the functional consequences of these aberrations. We propose that placental metabolic reprogramming reflects the dynamic physiological state allowing the tissue to adapt to developmental changes and respond to preeclampsia stress, whereas the inability to reprogram placental metabolism may result in severe preeclampsia phenotypes. Finally, we discuss common tested and novel therapeutic strategies for treating placental dysfunction in preeclampsia and their impact on placental energy metabolism as possible explanations into their potential benefits or harm.
Collapse
|
42
|
Dietrich B, Haider S, Meinhardt G, Pollheimer J, Knöfler M. WNT and NOTCH signaling in human trophoblast development and differentiation. Cell Mol Life Sci 2022; 79:292. [PMID: 35562545 PMCID: PMC9106601 DOI: 10.1007/s00018-022-04285-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/25/2022] [Accepted: 04/01/2022] [Indexed: 12/16/2022]
Abstract
Correct development of the human placenta and its differentiated epithelial cells, syncytial trophoblasts (STBs) and extravillous trophoblasts (EVTs), is crucial for a successful pregnancy outcome. STBs develop by cell fusion of mononuclear cytotrophoblasts (CTBs) in placental floating villi, whereas migratory EVTs originate from specialized villi anchoring to the maternal decidua. Defects in trophoblast differentiation have been associated with severe pregnancy disorders such as early-onset preeclampsia and fetal growth restriction. However, the evolutionary pathways underlying normal and adverse placentation are poorly understood. Herein, we discuss Wingless (WNT) and NOTCH signaling, two pathways that play pivotal roles in human placenta and trophoblast development. Whereas WNT is necessary for expansion of trophoblast progenitors and stem cells, NOTCH1 is required for proliferation and survival of EVT precursors. Differentiation of the latter is orchestrated by a switch in NOTCH receptor expression as well as by changes in WNT ligands and their downstream effectors.
Collapse
Affiliation(s)
- Bianca Dietrich
- grid.22937.3d0000 0000 9259 8492Placental Development Group, Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Währinger Gürtel 18–20, 5Q, 1090 Vienna, Austria
| | - Sandra Haider
- grid.22937.3d0000 0000 9259 8492Placental Development Group, Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Währinger Gürtel 18–20, 5Q, 1090 Vienna, Austria
| | - Gudrun Meinhardt
- grid.22937.3d0000 0000 9259 8492Placental Development Group, Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Währinger Gürtel 18–20, 5Q, 1090 Vienna, Austria
| | - Jürgen Pollheimer
- grid.22937.3d0000 0000 9259 8492Maternal-Fetal Immunology Group, Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Währinger Gürtel 18–20, 5Q, 1090 Vienna, Austria
| | - Martin Knöfler
- grid.22937.3d0000 0000 9259 8492Placental Development Group, Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Währinger Gürtel 18–20, 5Q, 1090 Vienna, Austria
| |
Collapse
|
43
|
Association between Paternal age and subtypes of preterm birth: A retrospective study. Reprod Biomed Online 2021; 44:1101-1109. [DOI: 10.1016/j.rbmo.2021.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 12/11/2021] [Accepted: 12/13/2021] [Indexed: 10/19/2022]
|
44
|
Oxidative stress-induced impairment of trophoblast function causes preeclampsia through the unfolded protein response pathway. Sci Rep 2021; 11:18415. [PMID: 34531444 PMCID: PMC8446002 DOI: 10.1038/s41598-021-97799-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 08/30/2021] [Indexed: 11/30/2022] Open
Abstract
Pre-eclampsia (PE) is a pregnancy-specific disorder, characterized by hypertension and proteinuria. In PE, trophoblasts mediated inadequate remodeling of uterine spiral arteries seem to interrupt uteroplacental blood flow, one of the hallmarks in the early onset of PE (EO-PE). This, in turn, results in placental ischemia–reperfusion injury during hypoxia and reoxygenation episodes, leading to the generation of reactive oxygen species (ROS) and oxidative stress (OS). But still it is debatable if OS is a cause or consequence of PE. In this present study, we have investigated the effects of OS on PE placentae and trophoblast cell functions using BeWo and HTR8/SVneo cell lines. PE placental tissues showed abnormal ultrastructure, high level of reactive oxygen species (ROS) with altered unfolded protein responses (UPR) in compare with term placental tissues. Similar to PE placentae, during OS induction, the trophoblast cells showed altered invasion and migration properties with significantly variable expression of differentiation and invasion markers, e.g., syncytin and MMPs. The effect was rescued by antioxidant, N-acetyl cysteine, thereby implying a ROS-specific effect and in the trophoblast cells, OS triggers UPR pathway through IRE1α-XBP1 axis. Taken together, these findings highlight the harmful effect of unfolded protein response, which was induced due to OS on trophoblast cells and deformed invasion and differentiation programme and can be extended further to clinical settings to identify clinically approved antioxidants during pregnancy as a therapeutic measure to reduce the onset of PE.
Collapse
|
45
|
Capatina N, Hemberger M, Burton GJ, Watson ED, Yung HW. Excessive endoplasmic reticulum stress drives aberrant mouse trophoblast differentiation and placental development leading to pregnancy loss. J Physiol 2021; 599:4153-4181. [PMID: 34269420 DOI: 10.1113/jp281994] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 07/05/2021] [Indexed: 12/17/2022] Open
Abstract
KEY POINTS Endoplasmic reticulum (ER) stress promotes placental dysmorphogenesis and is associated with poor pregnancy outcomes. We show that unfolded protein response signalling pathways located in the ER drive differentiation of mouse trophoblast stem cells into trophoblast subtypes involved in development of the placental labyrinth zone and trophoblast invasion. In a mouse model of chronic ER stress (Eif2s1tm1RjK ), higher ER stress in homozygous blastocysts is accompanied by reduced trophectoderm cell number and developmental delay and also is associated with an increased incidence of early pregnancy loss. Administration of the chemical chaperone, tauroursodeoxycholic acid, to Eif2s1+/ tm1RjK heterozygous females during pregnancy alleviated ER stress in the mutant placenta, restored normal trophoblast populations and reduced the frequency of early pregnancy loss. Our results suggest that alleviation of intrauterine ER stress could provide a potential therapeutic target to improve pregnancy outcome in women with pre-gestational metabolic or gynaecological conditions. ABSTRACT Women with pre-gestational health conditions (e.g. obesity, diabetes) or gynaecological problems (e.g. endometriosis) are at increased risk of adverse pregnancy outcomes including miscarriage, pre-eclampsia and fetal growth restriction. Increasing evidence suggests that unfavourable intrauterine conditions leading to poor implantation and/or defective placentation are a possible causative factor. The endoplasmic reticulum (ER) unfolded protein response (UPRER ) signalling pathways are a convergence point of various physiological stress stimuli that can be triggered by an unfavourable intrauterine environment. Therefore, we explored the impact of ER stress on mouse trophoblast differentiation in vitro, mouse blastocyst formation and early placenta development in the Eif2s1tm1RjK mutant mouse model of chronic ER stress. Chemically-manipulated ER stress or activation of UPRER pathways in a mouse trophoblast stem cell line promoted lineage-specific differentiation. Co-treatment with specific UPRER pathway inhibitors rescued this effect. Although the inner cell mass was unaffected, the trophectoderm of homozygous Eif2s1tm1RjK blastocysts exhibited ER stress associated with a reduced cell number. Furthermore, one-third of Eif2s1tm1RjK homozygous blastocysts exhibited severe developmental defects. We have previously reported a reduced trophoblast population and premature trophoblast differentiation in Eif2s1tm1RjK homozygous placentas at mid-gestation. Here, we demonstrate that treatment of Eif2s1+/tm1RjK heterozygous pregnant females with the chemical chaperone tauroursodeoxycholic acid alleviated ER stress, restored the trophoblast population and reduced the frequency of embryonic lethality. Our data suggest that therapeutic targeting of ER stress may improve pregnancy outcome in women with pre-gestational metabolic or gynaecological conditions.
Collapse
Affiliation(s)
- Nadejda Capatina
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Myriam Hemberger
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.,Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Graham J Burton
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Erica D Watson
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Hong Wa Yung
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
46
|
Warner GR, Dettogni RS, Bagchi IC, Flaws JA, Graceli JB. Placental outcomes of phthalate exposure. Reprod Toxicol 2021; 103:1-17. [PMID: 34015474 PMCID: PMC8260441 DOI: 10.1016/j.reprotox.2021.05.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/14/2021] [Accepted: 05/05/2021] [Indexed: 12/11/2022]
Abstract
Proper placental development and function relies on hormone receptors and signaling pathways that make the placenta susceptible to disruption by endocrine disrupting chemicals, such as phthalates. Here, we review relevant research on the associations between phthalate exposures and dysfunctions of the development and function of the placenta, including morphology, physiology, and genetic and epigenetic effects. This review covers in vitro studies, in vivo studies in mammals, and studies in humans. We also discuss important gaps in the literature. Overall, the evidence indicates that toxicity to the placental and maternal-fetal interface is associated with exposure to phthalates. Further studies are needed to better elucidate the mechanisms through which phthalates act in the placenta as well as additional human studies that assess placental disruption through pregnancy with larger sample sizes.
Collapse
Affiliation(s)
- Genoa R Warner
- Dept of Comparative Biosciences, University of Illinois, Urbana, IL, USA
| | | | - Indrani C Bagchi
- Dept of Comparative Biosciences, University of Illinois, Urbana, IL, USA
| | - Jodi A Flaws
- Dept of Comparative Biosciences, University of Illinois, Urbana, IL, USA.
| | - Jones B Graceli
- Dept of Morphology, Federal University of Espirito Santo, Brazil
| |
Collapse
|
47
|
Abstract
Tracing the early paths leading to developmental disorders is critical for prevention. In previous work, we detected an interaction between genomic risk scores for schizophrenia (GRSs) and early-life complications (ELCs), so that the liability of the disorder explained by genomic risk was higher in the presence of a history of ELCs, compared with its absence. This interaction was specifically driven by loci harboring genes highly expressed in placentae from normal and complicated pregnancies [G. Ursini et al., Nat. Med. 24, 792-801 (2018)]. Here, we analyze whether fractionated genomic risk scores for schizophrenia and other developmental disorders and traits, based on placental gene-expression loci (PlacGRSs), are linked with early neurodevelopmental outcomes in individuals with a history of ELCs. We found that schizophrenia's PlacGRSs are negatively associated with neonatal brain volume in singletons and offspring of multiple pregnancies and, in singletons, with cognitive development at 1 y and, less strongly, at 2 y, when cognitive scores become more sensitive to other factors. These negative associations are stronger in males, found only with GRSs fractionated by placental gene expression, and not found in PlacGRSs for other developmental disorders and traits. The relationship of PlacGRSs with brain volume persists as an anlage of placenta biology in adults with schizophrenia, again selectively in males. Higher placental genomic risk for schizophrenia, in the presence of ELCs and particularly in males, alters early brain growth and function, defining a potentially reversible neurodevelopmental path of risk that may be unique to schizophrenia.
Collapse
|
48
|
Heusler I, Biron-Shental T, Farladansky-Gershnabel S, Pasternak Y, Kidron D, Vulih-Shuitsman I, Einbinder Y, Cohen-Hagai K, Benchetrit S, Zitman-Gal T. Enhanced expression of Galectin-3 in gestational diabetes. Nutr Metab Cardiovasc Dis 2021; 31:1791-1797. [PMID: 34023181 DOI: 10.1016/j.numecd.2021.03.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/01/2021] [Accepted: 03/01/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS Gestational diabetes mellitus (GDM), hyperglycemia diagnosed during pregnancy, is one of the most common medical complications of pregnancy, treated primarily by diet and pharmacotherapy, if indicated. It is well-established that GDM increases the risk of adverse pregnancy outcomes and long-term complications in mothers and infants. Galectin-3 (Gal-3) is important in processes of cell growth, differentiation, inflammation, and fibrosis. We evaluated Gal-3 expression in pregnancies complicated by GDM as a parameter that might explain how GDM influences early onset of future complications. METHODS AND RESULTS Forty-four women with GDM and 40 with normal pregnancy (NP) were recruited during delivery admission. Blood samples were obtained from parturients and umbilical cords blood, as well as placental tissue for analysis. Gal-3 mRNA expression was increased in maternal blood samples and placental tissue of women with GDM compared to NP. In GDM, Gal-3 mRNA was decreased in cord blood compared to maternal blood and placental tissue. Gal-3 GDM placental protein expression was increased compared to NP. Immunostaining revealed that Gal-3 is upregulated in GDM placental extravillous trophoblast. ELISA of Gal-3 maternal serum levels between GDM and NP were similar. CONCLUSION Gal-3 is strongly expressed at molecular levels (mRNA and protein expression) in GDM maternal blood and placental tissue, and decreased in cord blood. These findings highlight the role of the placenta in protecting the fetus from potential Gal-3 damage. Gal-3 expression at mRNA and protein levels might be influenced by diabetes, even if blood glucose is balanced by medication or diet.
Collapse
Affiliation(s)
- Ishai Heusler
- Department of Obstetrics and Gynecology, Meir Medical Center, Kfar Saba, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tal Biron-Shental
- Department of Obstetrics and Gynecology, Meir Medical Center, Kfar Saba, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sivan Farladansky-Gershnabel
- Department of Obstetrics and Gynecology, Meir Medical Center, Kfar Saba, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yael Pasternak
- Department of Obstetrics and Gynecology, Meir Medical Center, Kfar Saba, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Debora Kidron
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Pathology Department, Meir Medical Center, Kfar Saba, Israel
| | | | - Yael Einbinder
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Nephrology Laboratory, Department of Nephrology and Hypertension, Meir Medical Center, Kfar Saba, Israel
| | - Keren Cohen-Hagai
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Nephrology Laboratory, Department of Nephrology and Hypertension, Meir Medical Center, Kfar Saba, Israel
| | - Sydney Benchetrit
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Nephrology Laboratory, Department of Nephrology and Hypertension, Meir Medical Center, Kfar Saba, Israel
| | - Tali Zitman-Gal
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Nephrology Laboratory, Department of Nephrology and Hypertension, Meir Medical Center, Kfar Saba, Israel.
| |
Collapse
|
49
|
Zhou J, West RC, Ehlers EL, Ezashi T, Schulz LC, Roberts RM, Yuan Y, Schust DJ. Modeling human peri-implantation placental development and function†. Biol Reprod 2021; 105:40-51. [PMID: 33899095 DOI: 10.1093/biolre/ioab080] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/16/2021] [Accepted: 04/20/2021] [Indexed: 12/17/2022] Open
Abstract
It is very difficult to gain a better understanding of the events in human pregnancy that occur during and just after implantation because such pregnancies are not yet clinically detectable. Animal models of human placentation are inadequate. In vitro models that utilize immortalized cell lines and cells derived from trophoblast cancers have multiple limitations. Primary cell and tissue cultures often have limited lifespans and cannot be obtained from the peri-implantation period. We present here two contemporary models of human peri-implantation placental development: extended blastocyst culture and stem-cell derived trophoblast culture. We discuss current research efforts that employ these models and how such models might be used in the future to study the "black box" stage of human pregnancy.
Collapse
Affiliation(s)
- J Zhou
- Mizzou Institute for Women's Health Research, Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, Columbia, MO USA.,Bond Life Sciences Center, Division of Animal Sciences, University of Missouri, Columbia, MO USA
| | - R C West
- Colorado Center for Reproductive Medicine, Lone Tree, CO USA
| | - E L Ehlers
- Mizzou Institute for Women's Health Research, Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, Columbia, MO USA
| | - T Ezashi
- Bond Life Sciences Center, Division of Animal Sciences, University of Missouri, Columbia, MO USA
| | - L C Schulz
- Mizzou Institute for Women's Health Research, Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, Columbia, MO USA
| | - R M Roberts
- Bond Life Sciences Center, Division of Animal Sciences, University of Missouri, Columbia, MO USA
| | - Y Yuan
- Colorado Center for Reproductive Medicine, Lone Tree, CO USA
| | - D J Schust
- Mizzou Institute for Women's Health Research, Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, Columbia, MO USA
| |
Collapse
|
50
|
Chen X, Guo DY, Yin TL, Yang J. Non-Coding RNAs Regulate Placental Trophoblast Function and Participate in Recurrent Abortion. Front Pharmacol 2021; 12:646521. [PMID: 33967782 PMCID: PMC8100504 DOI: 10.3389/fphar.2021.646521] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Recurrent spontaneous abortion (RSA) is a serious pregnancy complication with an increasing clinical incidence. The various causes of recurrent abortion are complicated. Developments in genetics, immunology, and cell biology have identified important roles of non-coding RNAs (ncRNAs) in the occurrence and progress of recurrent abortion. NcRNAs can affect the growth, migration, and invasion of placental trophoblasts by regulating cell processes such as the cell cycle, apoptosis, and epithelial-mesenchymal transformation. Therefore, their abnormal expression might lead to the occurrence and development of RSA. NcRNAs include small nuclear RNA (snRNA), small nucleolar RNA (snoRNA), ribosomal RNA (rRNA), transfer, RNA (tRNA), circular RNA (cRNA), and Piwi-interacting RNA (piRNA). In this review, we discuss recent research that focused on the function and mechanism of microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNA (circRNA) in regulating placental trophoblasts. The use of ncRNAs as potential diagnostic and predictive biomarkers in RSA is also discussed to provide future research insights.
Collapse
Affiliation(s)
- Xin Chen
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| | - Duan-Ying Guo
- Department of Gynecology, Longgang District People's Hospital of Shenzhen, Shenzhen, China
| | - Tai-Lang Yin
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| | - Jing Yang
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| |
Collapse
|