1
|
Su Y, Long J, Diao J, Li W, Chen X, Liao J, Tong C, Tan L, Zhang S, Li F, He J, Wang Y, Li C, Gao R. Dysregulation of the circ-Hdac4/miR-30c/RBPJ axis in decidua impairs placental function in preeclampsia. Cell Biol Toxicol 2025; 41:68. [PMID: 40208437 PMCID: PMC11985660 DOI: 10.1007/s10565-025-10016-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 03/25/2025] [Indexed: 04/11/2025]
Abstract
Embryo implantation relies on complex mother-fetus interactions. Abnormal decidualization can cause various pregnancy complications such as placental abnormalities, preeclampsia, and fetal growth restriction. circRNAs play a key role in various cellular processes. This study focuses on the role of circ-Hdac4, a circRNA derived from the Hdac4 gene, in decidualization and placental function. Mouse models revealed a spatiotemporally regulated expression of circ-Hdac4 in the endometrium during early pregnancy, with enhanced expression surrounding implantation sites. In vitro and in vivo assays confirmed that circ-Hdac4 is crucial for stromal cell decidualization, as its knockdown resulted in reduced expression of decidualization markers and disrupted endometrial architecture. Furthermore, we found that circ-Hdac4 functions as a microRNA sponge for miR-30c, which negatively regulates RBPJ, a critical protein for decidual remodeling. Proteomic analysis revealed that RBPJ was downregulated upon circ-Hdac4 silencing, and we validated the direct interaction between miR-30c and RBPJ using luciferase reporter assays. A mouse preeclampsia model showed that downregulation of circ-Hdac4 during decidualization exacerbated preeclampsia-related phenotypes, including reduced fetal counts, weights, and placental weights. In addition, we observed decreased expression of circ-Hdac4 and RBPJ in the decidual surface of placental tissues from preeclampsia patients, further supporting our findings in the mouse model. Collectively, our study provides evidence that circ-Hdac4 regulates decidualization through the miR-30c-RBPJ axis and that its abnormal expression during decidualization contributes to placental dysfunction in preeclampsia. This research offers novel insights into the molecular mechanisms underlying pregnancy complications and potential therapeutic targets for their prevention and treatment.
Collapse
Affiliation(s)
- Yan Su
- Joint International Research Laboratory of Reproduction & Development (Ministry of Education), School of Public Health, Chongqing Medical University, Chongqing, 400016, P. R. China
- School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
- Department of Clinical Laboratory, Chongqing Health Center for Women and Children/Women and Children's Hospital of Chongqing Medical University, Chongqing, 401147, P. R. China
| | - Jing Long
- Joint International Research Laboratory of Reproduction & Development (Ministry of Education), School of Public Health, Chongqing Medical University, Chongqing, 400016, P. R. China
- School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Jiani Diao
- Joint International Research Laboratory of Reproduction & Development (Ministry of Education), School of Public Health, Chongqing Medical University, Chongqing, 400016, P. R. China
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Weike Li
- Joint International Research Laboratory of Reproduction & Development (Ministry of Education), School of Public Health, Chongqing Medical University, Chongqing, 400016, P. R. China
- School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Xuemei Chen
- Joint International Research Laboratory of Reproduction & Development (Ministry of Education), School of Public Health, Chongqing Medical University, Chongqing, 400016, P. R. China
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Jiujiang Liao
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children/Women and Children's Hospital of Chongqing Medical University, Chongqing, 401147, P. R. China
| | - Chao Tong
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Liping Tan
- Joint International Research Laboratory of Reproduction & Development (Ministry of Education), School of Public Health, Chongqing Medical University, Chongqing, 400016, P. R. China
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Shuang Zhang
- Joint International Research Laboratory of Reproduction & Development (Ministry of Education), School of Public Health, Chongqing Medical University, Chongqing, 400016, P. R. China
- School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Fangfang Li
- Joint International Research Laboratory of Reproduction & Development (Ministry of Education), School of Public Health, Chongqing Medical University, Chongqing, 400016, P. R. China
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Junlin He
- Joint International Research Laboratory of Reproduction & Development (Ministry of Education), School of Public Health, Chongqing Medical University, Chongqing, 400016, P. R. China
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Yingxiong Wang
- Joint International Research Laboratory of Reproduction & Development (Ministry of Education), School of Public Health, Chongqing Medical University, Chongqing, 400016, P. R. China.
- School of Basic Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China.
| | - Chunli Li
- Joint International Research Laboratory of Reproduction & Development (Ministry of Education), School of Public Health, Chongqing Medical University, Chongqing, 400016, P. R. China.
- Department of Clinical Laboratory, Chongqing Health Center for Women and Children/Women and Children's Hospital of Chongqing Medical University, Chongqing, 401147, P. R. China.
| | - Rufei Gao
- Joint International Research Laboratory of Reproduction & Development (Ministry of Education), School of Public Health, Chongqing Medical University, Chongqing, 400016, P. R. China.
- Department of Health Toxicology, School of Public Health, Chongqing Medical University, Chongqing, 400016, P. R. China.
- Department of Obstetrics and Gynecology, Chongqing Health Center for Women and Children/Women and Children's Hospital of Chongqing Medical University, Chongqing, 401147, P. R. China.
| |
Collapse
|
2
|
Van Schoor K, Bruet E, Jones EAV, Migeotte I. Origin and flow-mediated remodeling of the murine and human extraembryonic circulation systems. Front Physiol 2024; 15:1395006. [PMID: 38818524 PMCID: PMC11137303 DOI: 10.3389/fphys.2024.1395006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 04/16/2024] [Indexed: 06/01/2024] Open
Abstract
The transduction of mechanical stimuli produced by blood flow is an important regulator of vascular development. The vitelline and umbilico-placental circulations are extraembryonic vascular systems that are required for proper embryonic development in mammalian embryos. The morphogenesis of the extraembryonic vasculature and the cardiovascular system of the embryo are hemodynamically and molecularly connected. Here we provide an overview of the establishment of the murine and human vitelline and umbilico-placental vascular systems and how blood flow influences various steps in their development. A deeper comprehension of extraembryonic vessel development may aid the establishment of stem-cell based embryo models and provide novel insights to understanding pregnancy complications related to the umbilical cord and placenta.
Collapse
Affiliation(s)
- Kristof Van Schoor
- Institut de Recherche Interdisciplinaire Jacques E. Dumont, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Emmanuel Bruet
- Institut de Recherche Interdisciplinaire Jacques E. Dumont, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Elizabeth Anne Vincent Jones
- Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
- Department of Cardiology CARIM School for Cardiovascular Diseases Maastricht University, Maastricht, Netherlands
| | - Isabelle Migeotte
- Institut de Recherche Interdisciplinaire Jacques E. Dumont, Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
3
|
Zhang X, He H, Yu H, Teng X, Wang Z, Li C, Li J, Yang H, Shen J, Wu T, Zhang F, Zhang Y, Wu Q. Maternal RNA transcription in Dlk1-Dio3 domain is critical for proper development of the mouse placental vasculature. Commun Biol 2024; 7:363. [PMID: 38521877 PMCID: PMC10960817 DOI: 10.1038/s42003-024-06038-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 03/11/2024] [Indexed: 03/25/2024] Open
Abstract
The placenta is a unique organ for ensuring normal embryonic growth in the uterine. Here, we found that maternal RNA transcription in Dlk1-Dio3 imprinted domain is essential for placentation. PolyA signals were inserted into Gtl2 to establish a mouse model to prevent the expression of maternal RNAs in the domain. The maternal allele knock-in (MKI) and homozygous (HOMO) placentas showed an expanded junctional zone, reduced labyrinth and poor vasculature impacting both fetal and maternal blood spaces. The MKI and HOMO models displayed dysregulated gene expression in the Dlk1-Dio3 domain. In situ hybridization detected Dlk1, Gtl2, Rtl1, miR-127 and Rian dysregulated in the labyrinth vasculature. MKI and HOMO induced Dlk1 to lose imprinting, and DNA methylation changes of IG-DMR and Gtl2-DMR, leading to abnormal gene expression, while the above changes didn't occur in paternal allele knock-in placentas. These findings demonstrate that maternal RNAs in the Dlk1-Dio3 domain are involved in placental vasculature, regulating gene expression, imprinting status and DNA methylation.
Collapse
Affiliation(s)
- Ximeijia Zhang
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, Heilongjiang, China
| | - Hongjuan He
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, Heilongjiang, China
| | - Haoran Yu
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, Heilongjiang, China
| | - Xiangqi Teng
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, Heilongjiang, China
| | - Ziwen Wang
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, Heilongjiang, China
| | - Chenghao Li
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, Heilongjiang, China
| | - Jiahang Li
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, Heilongjiang, China
| | - Haopeng Yang
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, Heilongjiang, China
| | - Jiwei Shen
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, Heilongjiang, China
| | - Tong Wu
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, Heilongjiang, China
| | - Fengwei Zhang
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, Heilongjiang, China
| | - Yan Zhang
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, Heilongjiang, China
| | - Qiong Wu
- School of Life Science and Technology, State Key Laboratory of Urban Water Resource and Environment, Harbin Institute of Technology, Harbin, 150006, Heilongjiang, China.
| |
Collapse
|
4
|
Xin Q, Yu G, Feng I, Dean J. Chromatin remodeling of prostaglandin signaling in smooth muscle enables mouse embryo passage through the female reproductive tract. Dev Cell 2023; 58:1716-1732.e8. [PMID: 37714160 DOI: 10.1016/j.devcel.2023.08.025] [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: 02/14/2023] [Revised: 05/10/2023] [Accepted: 08/22/2023] [Indexed: 09/17/2023]
Abstract
Early mammalian development occurs during embryo transit of the female reproductive tract. Transport is orchestrated by secreted oviduct fluid, unidirectional beating of epithelial cilia, and smooth muscle contractions. Using gene-edited mice, we document that conditional disruption of a component of the SWI/SNF chromatin remodeling complex in smooth muscle cells prevents transport through the oviduct without perturbing embryogenesis. Analysis with RNA sequencing (RNA-seq), transposase-accessible chromatin with sequencing (ATAC-seq), chromatin immunocleavage sequencing (ChIC-seq), and pharmacologic rescue experiments implicated prostaglandin signaling pathways. In comparison with controls, gene-edited mice had compromised chromatin accessibility at enhancer/promoters of Ptgs2, Pla2g16, Pla2r1, and Ptger3 (EP3) as well as decreased enhancer-promoter interactive looping critical for Ptgs2 (aka Cox-2) expression in a SWI/SNF complex-dependent manner. Treatment of wild-type mice with prostaglandin inhibitors phenocopied the genetically induced defect.
Collapse
Affiliation(s)
- Qiliang Xin
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Guoyun Yu
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Iris Feng
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jurrien Dean
- Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
5
|
Anamthathmakula P, Shallie PD, Nayak N, Dhal S, Vivian JL, Mor G, Soares MJ, Nayak NR. Variable Cre Recombination Efficiency in Placentas of Cyp19-Cre ROSA mT/mG Transgenic Mice. Cells 2023; 12:2096. [PMID: 37626906 PMCID: PMC10453067 DOI: 10.3390/cells12162096] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/06/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
The aromatase-Cre recombinase (Cyp19-Cre) transgenic mouse model has been extensively used for placenta-specific gene inactivation. In a pilot study, we observed unexpected phenotypes using this mouse strain, which prompted an extensive characterization of Cyp19-Cre placental phenotypes using ROSAmT/mG transgenic reporter mice. The two strains were mated to generate bi-transgenic Cyp19-Cre;ROSAmT/mG mice following a standard transgenic breeding scheme, and placental and fetal tissues were analyzed on embryonic day 17.5. Both maternal and paternal Cre inheritance were analyzed by mating the respective Cyp19-Cre and ROSAmT/mG males and females. The genotype results showed the expected percentage of Cyp19-Cre;ROSAmT/mG fetuses (73%) and Cre mRNA was expressed in all of the Cyp19-Cre placentas. However, surprisingly, only about 50% of the Cyp19-Cre;ROSAmT/mG placentas showed Cre-mediated recombinase activity as demonstrated by placental enhanced green fluorescent protein (EGFP) expression. Further genetic excision analysis of the placentas revealed consistent results showing the absence of excision of the tdTomato in all of the Cyp19-Cre;ROSAmT/mG placentas lacking EGFP expression. Moreover, among the EGFP-expressing placentas, there was wide variability in recombination efficiency, even in placentas from the same litter, leading to a mosaic pattern of EGFP expression in different zones and cell types of the placentas. In addition, we observed a significantly higher percentage of Cre recombination activity in placentas with maternal Cre inheritance. Our results show frequent mosaicism, inconsistent recombination activity, and parent-of-origin effects in placentas from Cyp19-Cre;ROSAmT/mG mice, suggesting that tail-biopsy genotype results may not necessarily indicate the excision of floxed genes in Cyp19-Cre positive placentas. Thus, placenta-specific mutagenesis studies using the Cyp19-Cre model require extensive characterization and careful interpretation of the placental phenotypes for each floxed allele.
Collapse
Affiliation(s)
- Prashanth Anamthathmakula
- Department of Obstetrics and Gynecology, University of Missouri-Kansas City School of Medicine, Kansas City, MO 64108, USA
- Department of Surgery, University of Missouri Kansas City School of Medicine, Kansas City, MO 64108, USA
| | - Philemon D. Shallie
- Department of Obstetrics and Gynecology, University of Missouri-Kansas City School of Medicine, Kansas City, MO 64108, USA
- Department of Surgery, University of Missouri Kansas City School of Medicine, Kansas City, MO 64108, USA
| | - Neha Nayak
- Department of Obstetrics and Gynecology, University of Missouri-Kansas City School of Medicine, Kansas City, MO 64108, USA
| | - Sabita Dhal
- Department of Obstetrics and Gynecology, University of Missouri-Kansas City School of Medicine, Kansas City, MO 64108, USA
| | - Jay L. Vivian
- Children’s Mercy Research Institute, Children’s Mercy, Kansas City, MO 64108, USA
| | - Gil Mor
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
| | - Michael J. Soares
- Institute for Reproductive and Developmental Sciences, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Center for Perinatal Research, Children’s Mercy Research Institute, Children’s Mercy, Kansas City, MO 64108, USA
| | - Nihar R. Nayak
- Department of Obstetrics and Gynecology, University of Missouri-Kansas City School of Medicine, Kansas City, MO 64108, USA
| |
Collapse
|
6
|
Moldovan GE, Miele L, Fazleabas AT. Notch signaling in reproduction. Trends Endocrinol Metab 2021; 32:1044-1057. [PMID: 34479767 PMCID: PMC8585702 DOI: 10.1016/j.tem.2021.08.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/27/2021] [Accepted: 08/06/2021] [Indexed: 12/22/2022]
Abstract
The Notch signaling pathway is conserved among mammalian species and controls proliferation, differentiation, and cell death in many organs throughout the body including the reproductive tract. Notch signaling plays critical roles in the development and function of both the male and female reproductive systems. Specifically, within the female reproductive tract, Notch signaling is hormone regulated and mediates key reproductive events important for ovarian and uterine function. In this review, we highlight the tissues that express Notch receptors, ligands, and downstream effectors and distinguish how these molecules regulate reproductive function in male and female mice, non-human primates, and humans. Finally, we describe some of the aberrations in Notch signaling in female reproductive pathologies and identify opportunities for future investigation.
Collapse
Affiliation(s)
- Genna E Moldovan
- Department of Obstetrics, Gynecology, and Reproductive Biology, Michigan State University, Grand Rapids, MI 49503, USA
| | - Lucio Miele
- Department of Genetics, Louisiana State University Health Sciences Center and Stanley S. Scott Cancer Center, New Orleans, LA 70112, USA
| | - Asgerally T Fazleabas
- Department of Obstetrics, Gynecology, and Reproductive Biology, Michigan State University, Grand Rapids, MI 49503, USA.
| |
Collapse
|
7
|
Starks RR, Kaur H, Tuteja G. Mapping cis-regulatory elements in the midgestation mouse placenta. Sci Rep 2021; 11:22331. [PMID: 34785717 PMCID: PMC8595355 DOI: 10.1038/s41598-021-01664-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 11/01/2021] [Indexed: 12/22/2022] Open
Abstract
The placenta is a temporary organ that provides the developing fetus with nutrients, oxygen, and protection in utero. Defects in its development, which may be caused by misregulated gene expression, can lead to devastating outcomes for the mother and fetus. In mouse, placental defects during midgestation commonly lead to embryonic lethality. However, the regulatory mechanisms controlling expression of genes during this period have not been thoroughly investigated. Therefore, we generated and analyzed ChIP-seq data for multiple histone modifications known to mark cis-regulatory regions. We annotated active and poised promoters and enhancers, as well as regions generally associated with repressed gene expression. We found that poised promoters were associated with neuronal development genes, while active promoters were largely associated with housekeeping genes. Active and poised enhancers were associated with placental development genes, though only active enhancers were associated with genes that have placenta-specific expression. Motif analysis within active enhancers identified a large network of transcription factors, including those that have not been previously studied in the placenta and are candidates for future studies. The data generated and genomic regions annotated provide researchers with a foundation for future studies, aimed at understanding how specific genes in the midgestation mouse placenta are regulated.
Collapse
Affiliation(s)
- Rebekah R Starks
- Genetics, Development, and Cell Biology, Iowa State University, Ames, IA, 50011, USA.,Bioinformatics and Computational Biology, Iowa State University, Ames, IA, 50011, USA
| | - Haninder Kaur
- Genetics, Development, and Cell Biology, Iowa State University, Ames, IA, 50011, USA
| | - Geetu Tuteja
- Genetics, Development, and Cell Biology, Iowa State University, Ames, IA, 50011, USA. .,Bioinformatics and Computational Biology, Iowa State University, Ames, IA, 50011, USA.
| |
Collapse
|
8
|
Perlman BE, Merriam AA, Lemenze A, Zhao Q, Begum S, Nair M, Wu T, Wapner RJ, Kitajewski JK, Shawber CJ, Douglas NC. Implications for preeclampsia: hypoxia-induced Notch promotes trophoblast migration. Reproduction 2021; 161:681-696. [PMID: 33784241 PMCID: PMC8403268 DOI: 10.1530/rep-20-0483] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 03/30/2021] [Indexed: 01/15/2023]
Abstract
In the first trimester of human pregnancy, low oxygen tension or hypoxia is essential for proper placentation and placenta function. Low oxygen levels and activation of signaling pathways have been implicated as critical mediators in the promotion of trophoblast differentiation, migration, and invasion with inappropriate changes in oxygen tension and aberrant Notch signaling both individually reported as causative to abnormal placentation. Despite crosstalk between hypoxia and Notch signaling in multiple cell types, the relationship between hypoxia and Notch in first trimester trophoblast function is not understood. To determine how a low oxygen environment impacts Notch signaling and cellular motility, we utilized the human first trimester trophoblast cell line, HTR-8/SVneo. Gene set enrichment and ontology analyses identified pathways involved in angiogenesis, Notch and cellular migration as upregulated in HTR-8/SVneo cells exposed to hypoxic conditions. DAPT, a γ-secretase inhibitor that inhibits Notch activation, was used to interrogate the crosstalk between Notch and hypoxia pathways in HTR-8/SVneo cells. We found that hypoxia requires Notch activation to mediate HTR-8/SVneo cell migration, but not invasion. To determine if our in vitro findings were associated with preeclampsia, we analyzed the second trimester chorionic villous sampling (CVS) samples and third trimester placentas. We found a significant decrease in expression of migration and invasion genes in CVS from preeclamptic pregnancies and significantly lower levels of JAG1 in placentas from pregnancies with early-onset preeclampsia with severe features. Our data support a role for Notch in mediating hypoxia-induced trophoblast migration, which may contribute to preeclampsia development.
Collapse
Affiliation(s)
- Barry E Perlman
- Department of Obstetrics, Gynecology and Reproductive Health, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Audrey A. Merriam
- Department of Obstetrics, Gynecology and Reproductive Sciences Yale University, New Haven, CT, USA
| | - Alexander Lemenze
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
- Center for Immunity and Inflammation, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Qingshi Zhao
- Department of Obstetrics, Gynecology and Reproductive Health, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Salma Begum
- Department of Obstetrics, Gynecology and Reproductive Health, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Mohan Nair
- Department of Obstetrics, Gynecology and Reproductive Health, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Tracy Wu
- Department of Obstetrics, Gynecology and Reproductive Health, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Ronald J. Wapner
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Jan K. Kitajewski
- Department of Physiology & Biophysics, University of Illinois Chicago, Chicago, IL, USA
| | - Carrie J. Shawber
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Nataki C. Douglas
- Department of Obstetrics, Gynecology and Reproductive Health, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
- Center for Immunity and Inflammation, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| |
Collapse
|
9
|
Y-Box Binding Protein 1 Expression in Trophoblast Cells Promotes Fetal and Placental Development. Cells 2020; 9:cells9091942. [PMID: 32842598 PMCID: PMC7563187 DOI: 10.3390/cells9091942] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/20/2020] [Accepted: 08/21/2020] [Indexed: 12/14/2022] Open
Abstract
Y-box binding protein 1 (YB-1) is pivotal for the regulation of cancerogenesis and inflammation. However, its involvement in pregnancy processes such as fetal and placental development remains to be elucidated. We studied Ybx1 (YB-1)+/− heterozygous intercrossings and compared them to YB-1+/+ wild-type (WT) combinations. Additionally, we generated trophoblast-specific YB-1-deficient mice by pairing FVB Cyp19-Cre females to YB-1fl/fl males. YB-1fl/fl-paired FVB WT females served as controls. Serial in vivo ultrasound measurements were performed to assess fetal and placental parameters. After sacrificing the females, implantation and abortion rates were recorded, spiral artery (SA) remodeling was analyzed and fetal and placental weights were determined. Compared to YB-1+/+ counterparts, YB-1+/− females showed reduced implantation areas at gestation day (GD)10, insufficiently remodeled SAs at GD12, increased placental diameter/thickness ratios at GD14 and reduced placental and fetal weights at GD14. Compared to WT, Cyp19-Cre females with YB-1-deficient placentas showed reduced implantation areas at GD8, 10 and 12; decreased placental areas and diameters at GD10 and 12; diminished placental thicknesses at GD12; as well as reduced placental weights at GD12 and 14. In conclusion, our data suggest haploinsufficiency of YB-1 resulting in disturbed fetal and placental development. Moreover, we provide the first evidence for the relevance of trophoblast-specific YB-1 for placentation.
Collapse
|