1
|
Li R, Ma L, Geng Y, Chen X, Zhu J, Zhu H, Wang D. Uteroplacental microvascular remodeling in health and disease. Acta Physiol (Oxf) 2025; 241:e70035. [PMID: 40156319 DOI: 10.1111/apha.70035] [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: 09/25/2024] [Revised: 02/10/2025] [Accepted: 03/13/2025] [Indexed: 04/01/2025]
Abstract
The microvascular system is essential for delivering oxygen and nutrients to tissues while removing metabolic waste. During pregnancy, the uteroplacental microvascular system undergoes extensive remodeling to meet the increased demands of the fetus. Key adaptations include vessel dilation and increases in vascular volume, density, and permeability, all of which ensure adequate placental perfusion while maintaining stable maternal blood pressure. Structural and functional abnormalities in the uteroplacental microvasculature are associated with various gestational complications, posing both immediate and long-term risks to the health of both mother and infant. In this review, we describe the changes in uteroplacental microvessels during pregnancy, discuss the pathogenic mechanisms underlying diseases such as preeclampsia, fetal growth restriction, and gestational diabetes, and summarize current clinical and research approaches for monitoring microvascular health. We also provide an update on research models for gestational microvascular complications and explore solutions to several unresolved challenges. With advancements in research techniques, we anticipate significant progress in understanding and managing these diseases, ultimately leading to new therapeutic strategies to improve maternal and fetal health.
Collapse
Affiliation(s)
- Ruizhi Li
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Qingdao University, Jinan, China
- Institute of Chronic Diseases, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Lei Ma
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Qingdao University, Jinan, China
- Institute of Chronic Diseases, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Yingchun Geng
- Institute of Chronic Diseases, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
- Department of Reproductive Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Xiaoxue Chen
- Institute of Chronic Diseases, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jiaxi Zhu
- Life Sciences, Faculty of Arts & Science, University of Toronto - St. George Campus, Toronto, Ontario, Canada
| | - Hai Zhu
- Department of Urology, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
- Department of Urology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Dong Wang
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Qingdao University, Jinan, China
- Institute of Chronic Diseases, The Affiliated Hospital of Qingdao University, Qingdao Medical College, Qingdao University, Qingdao, China
| |
Collapse
|
2
|
Li W, Zhang Q, Ni M, Li B, Chen Z, Shen Q, Lin Z, Cheng C, Yao D, Qi S, Ding X, Shen H, Liu X, Tang Z, Huang X, Zhao J, Liu Z. Upregulated YTHDC1 mediates trophoblastic dysfunction inducing preterm birth in ART conceptions through enhanced RPL37 translation. Cell Mol Life Sci 2024; 82:17. [PMID: 39725796 DOI: 10.1007/s00018-024-05467-x] [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: 05/21/2024] [Revised: 09/01/2024] [Accepted: 10/01/2024] [Indexed: 12/28/2024]
Abstract
Assisted reproductive technology (ART) pregnancies present a higher risk of singleton preterm birth than natural pregnancies, but the underlying molecular mechanism remains largely unknown. RNA m6A modification is a key epigenetic mechanism regulating cellular function, but the role of m6A modification, especially its "reader" YTHDC1, in preterm delivery remains undefined. To delineate the role and epigenetic mechanism of m6A modification in ART preterm delivery, the effects of YTHDC1 on trophoblastic function were evaluated by CCK-8, EdU, Transwell, and flow cytometry analyses post its overexpression or knockdown. Downstream signaling pathways of YTHDC1 were investigated by RNA-seq, and targeted mRNAs were explored by RIP-seq and MeRIP-seq. Upstream transcriptional factors of YTHDC1 were determined by ChIP-seq and luciferase reporter assays. Elevated YTHDC1 was detected in human ART-conceived preterm placentas and in murine preterm placentas post estradiol (E2) exposure. In vitro experiments showed that YTHDC1 promoted trophoblastic cell proliferation and migration, but inhibited cell apoptosis. Mechanistically, E2 was proven to upregulate YTHDC1 expression via retinoid X receptor alpha (RXRA) in trophoblastic cells. Enhanced YTHDC1 expression augmented the translation of RPL37 in an m6A-dependent manner by binding to m6A-modified RPL37 mRNA and concomitantly promoted the overall translational output. Importantly, administration of siRNA targeting YTHDC1 effectively delayed the progression of preterm delivery. In conclusion, the identified E2/RXRA/YTHDC1/RPL37 axis provides new insights into the epigenetic mechanism underlying ART-associated preterm delivery. The findings offer a potential prognostic biomarker and therapeutic target for preterm delivery.
Collapse
Affiliation(s)
- Wei Li
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Qianqian Zhang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Meng Ni
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Baihe Li
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Ze Chen
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Qianwen Shen
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Zhenying Lin
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Chunyu Cheng
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Dongting Yao
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Sudong Qi
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Xiya Ding
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Haiqing Shen
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
| | - Xiaorui Liu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Zheng Tang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
| | - Xiaoyi Huang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
| | - Jiuru Zhao
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China.
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China.
| | - Zhiwei Liu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China.
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China.
| |
Collapse
|
3
|
Azagury M, Buganim Y. Unlocking trophectoderm mysteries: In vivo and in vitro perspectives on human and mouse trophoblast fate induction. Dev Cell 2024; 59:941-960. [PMID: 38653193 DOI: 10.1016/j.devcel.2024.03.029] [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: 09/07/2023] [Revised: 11/10/2023] [Accepted: 03/18/2024] [Indexed: 04/25/2024]
Abstract
In recent years, the pursuit of inducing the trophoblast stem cell (TSC) state has gained prominence as a compelling research objective, illuminating the establishment of the trophoblast lineage and unlocking insights into early embryogenesis. In this review, we examine how advancements in diverse technologies, including in vivo time course transcriptomics, cellular reprogramming to TSC state, chemical induction of totipotent stem-cell-like state, and stem-cell-based embryo-like structures, have enriched our insights into the intricate molecular mechanisms and signaling pathways that define the mouse and human trophectoderm/TSC states. We delve into disparities between mouse and human trophectoderm/TSC fate establishment, with a special emphasis on the intriguing role of pluripotency in this context. Additionally, we re-evaluate recent findings concerning the potential of totipotent-stem-like cells and embryo-like structures to fully manifest the trophectoderm/trophoblast lineage's capabilities. Lastly, we briefly discuss the potential applications of induced TSCs in pregnancy-related disease modeling.
Collapse
Affiliation(s)
- Meir Azagury
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Yosef Buganim
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel.
| |
Collapse
|
4
|
Zhang Q, Tian Y, Fu Z, Wu S, Lan H, Zhou X, Shen W, Lou Y. The role of serum-glucocorticoid regulated kinase 1 in reproductive viability: implications from prenatal programming and senescence. Mol Biol Rep 2024; 51:376. [PMID: 38427115 PMCID: PMC10907440 DOI: 10.1007/s11033-024-09341-8] [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: 11/21/2023] [Accepted: 02/09/2024] [Indexed: 03/02/2024]
Abstract
OBJECTIVE Organisms and cellular viability are of paramount importance to living creatures. Disruption of the balance between cell survival and apoptosis results in compromised viability and even carcinogenesis. One molecule involved in keeping this homeostasis is serum-glucocorticoid regulated kinase (SGK) 1. Emerging evidence points to a significant role of SGK1 in cell growth and survival, cell metabolism, reproduction, and life span, particularly in prenatal programming and reproductive senescence by the same token. Whether the hormone inducible SGK1 kinase is a major driver in the pathophysiological processes of prenatal programming and reproductive senescence? METHOD The PubMed/Medline, Web of Science, Embase/Ovid, and Elsevier Science Direct literature databases were searched for articles in English focusing on SGK1 published up to July 2023 RESULT: Emerging evidence is accumulating pointing to a pathophysiological role of the ubiquitously expressed SGK1 in the cellular and organismal viability. Under the regulation of specific hormones, extracellular stimuli, and various signals, SGK1 is involved in several biological processes relevant to viability, including cell proliferation and survival, cell migration and differentiation. In line, SGK1 contributes to the development of germ cells, embryos, and fetuses, whereas SGK1 inhibition leads to abnormal gametogenesis, embryo loss, and truncated reproductive lifespan. CONCLUTION SGK1 integrates a broad spectrum of effects to maintain the homeostasis of cell survival and apoptosis, conferring viability to multiple cell types as well as both simple and complex organisms, and thus ensuring appropriate prenatal development and reproductive lifespan.
Collapse
Affiliation(s)
- Qiying Zhang
- Department of Gynaecology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No. 453 Tiyuchang Road, Hangzhou, 310007, Zhejiang, China
| | - Ye Tian
- Department of Gynaecology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No. 453 Tiyuchang Road, Hangzhou, 310007, Zhejiang, China
| | - Zhujing Fu
- Jinhua Municipal Central Hospital, Jinhua, 321001, China
| | - Shuangyu Wu
- Medical School, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Huizhen Lan
- Department of Gynaecology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No. 453 Tiyuchang Road, Hangzhou, 310007, Zhejiang, China
| | - Xuanle Zhou
- Department of Gynaecology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No. 453 Tiyuchang Road, Hangzhou, 310007, Zhejiang, China
| | - Wendi Shen
- Department of Gynaecology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No. 453 Tiyuchang Road, Hangzhou, 310007, Zhejiang, China
| | - Yiyun Lou
- Department of Gynaecology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No. 453 Tiyuchang Road, Hangzhou, 310007, Zhejiang, China.
| |
Collapse
|
5
|
Lin Z, Wu S, Jiang Y, Chen Z, Huang X, Wen Z, Yuan Y. Unraveling the molecular mechanisms driving enhanced invasion capability of extravillous trophoblast cells: a comprehensive review. J Assist Reprod Genet 2024; 41:591-608. [PMID: 38315418 PMCID: PMC10957806 DOI: 10.1007/s10815-024-03036-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/15/2024] [Indexed: 02/07/2024] Open
Abstract
Precise extravillous trophoblast (EVT) invasion is crucial for successful placentation and pregnancy. This review focuses on elucidating the mechanisms that promote heightened EVT invasion. We comprehensively summarize the pivotal roles of hormones, angiogenesis, hypoxia, stress, the extracellular matrix microenvironment, epithelial-to-mesenchymal transition (EMT), immunity, inflammation, programmed cell death, epigenetic modifications, and microbiota in facilitating EVT invasion. The molecular mechanisms underlying enhanced EVT invasion may provide valuable insights into potential pathogenic mechanisms associated with diseases characterized by excessive invasion, such as the placenta accreta spectrum (PAS), thereby offering novel perspectives for managing pregnancy complications related to deficient EVT invasion.
Collapse
Affiliation(s)
- Zihan Lin
- School of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Shuang Wu
- School of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Yinghui Jiang
- School of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Ziqi Chen
- School of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Xiaoye Huang
- School of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Zhuofeng Wen
- The Sixth Clinical School of Guangzhou Medical University, Guangzhou, China
| | - Yi Yuan
- School of Pediatrics, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
6
|
Ye R, Li Z, Xian H, Zhong Y, Liang B, Huang Y, Chen D, Dai M, Tang S, Guo J, Bai R, Feng Y, Chen Z, Yang X, Huang Z. Combined Effects of Polystyrene Nanosphere and Homosolate Exposures on Estrogenic End Points in MCF-7 Cells and Zebrafish. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:27011. [PMID: 38381479 PMCID: PMC10880820 DOI: 10.1289/ehp13696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 02/22/2024]
Abstract
BACKGROUND Micro- and nanoplastics (MNPs) and homosalate (HMS) are ubiquitous emerging environmental contaminants detected in human samples. Despite the well-established endocrine-disrupting effects (EDEs) of HMS, the interaction between MNPs and HMS and its impact on HMS-induced EDEs remain unclear. OBJECTIVES This study aimed to investigate the influence of MNPs on HMS-induced estrogenic effects and elucidate the underlying mechanisms in vitro and in vivo. METHODS We assessed the impact of polystyrene nanospheres (PNSs; 50 nm , 1.0 mg / L ) on HMS-induced MCF-7 cell proliferation (HMS: 0.01 - 1 μ M , equivalent to 2.62 - 262 μ g / L ) using the E-SCREEN assay and explored potential mechanisms through transcriptomics. Adult zebrafish were exposed to HMS (0.0262 - 262 μ g / L ) with or without PNSs (50 nm , 1.0 mg / L ) for 21 d. EDEs were evaluated through gonadal histopathology, fertility tests, steroid hormone synthesis, and gene expression changes in the hypothalamus-pituitary-gonad-liver (HPGL) axis. RESULTS Coexposure of HMS and PNSs resulted in higher expression of estrogen receptor α (ESR1) and the mRNAs of target genes (pS2, AREG, and PGR), a greater estrogen-responsive element transactivation activity, and synergistic stimulation on MCF-7 cell proliferation. Knockdown of serum and glucocorticoid-regulated kinase 1 (SGK1) rescued the MCF-7 cell proliferation induced by PNSs alone or in combination with HMS. In zebrafish, coexposure showed higher expression of SGK1 and promoted ovary development but inhibited spermatogenesis. In addition, coexposure led to lower egg hatchability, higher embryonic mortality, and greater larval malformation. Coexposure also modulated steroid hormone synthesis genes (cyp17a2, hsd17[Formula: see text]1, esr2b, vtg1, and vtg2), and resulted in higher 17 β -estradiol (E 2 ) release in females. Conversely, males showed lower testosterone, E 2 , and gene expressions of cyp11a1, cyp11a2, cyp17a1, cyp17a2, and hsd17[Formula: see text]1. DISCUSSION PNS exposure exacerbated HMS-induced estrogenic effects via SGK1 up-regulation in MCF-7 cells and disrupting the HPGL axis in zebrafish, with gender-specific patterns. This offers new mechanistic insights and health implications of MNP and contaminant coexposure. https://doi.org/10.1289/EHP13696.
Collapse
Affiliation(s)
- Rongyi Ye
- National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zhiming Li
- National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Hongyi Xian
- National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yizhou Zhong
- National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Boxuan Liang
- National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yuji Huang
- National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Da Chen
- College of Environment and Climate, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou, China
| | | | - Shuqin Tang
- College of Environment and Climate, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou, China
| | - Jie Guo
- Hunter Biotechnology, Inc, Hangzhou, China
| | - Ruobing Bai
- National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yu Feng
- National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zhenguo Chen
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xingfen Yang
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Food Safety and Health Research Center, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zhenlie Huang
- National Medical Products Administration (NMPA) Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou, China
| |
Collapse
|
7
|
Di X, Duan Z, Ma Y, Song X, Hao Y, Li G, Tan Z, Lou Y, Lin X. Jiawei Shoutai Pill promotes decidualization by regulating the SGK1/ENaC pathway in recurrent spontaneous abortion. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116939. [PMID: 37479068 DOI: 10.1016/j.jep.2023.116939] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/23/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jiawei Shoutai Pill (JWSTW) is a traditional herbal formula for recurrent spontaneous abortion (RSA). Although JWSTW significantly improves the clinical symptoms of RSA patients, its molecular mechanism remains unclear. AIM OF STUDY This study evaluated the expression and function of the serum/glucocorticoid regulated kinase 1/epithelial sodium channel (SGK1/ENaC) pathway and decidualization level in RSA patients and mice. It also investigated the therapeutic effects and potential mechanisms of JWSTW. MATERIALS AND METHODS 30 early RSA patients and 30 normal pregnant women undergoing induced abortion during the same period were included in the study. Decidual tissues were collected, and HE staining, immunohistochemistry, Western blot, and RT-PCR were used to detect protein and mRNA expression levels of SGK1, ENaC-a, estrogen Rreceptor β (ERβ), and progesterone receptor (PR) in patients' decidual tissues. Protein expression levels of prolactin receptor (PRLR) and insulin-like growth factor binding protein 1 (IGFBP-1) were also detected. A classical RSA mouse model was constructed, and the mice were randomly divided into four groups: normal, model, dydrogesterone (DQYT) (0.33 g/kg/d), and JWSTW (1.66 g/kg/d). The normal and model groups received the same volume of distilled water by gavage for 8 and 14 days after pregnancy. On the 14th day of pregnancy, the embryonic loss rate of each group, the number of offspring born to naturally delivered mice, and the protein or mRNA expression levels of key factors of the SGK1/ENaC pathway (SGK1, ENaC-a, ERβ, and PR), decidual proliferation marker (Ki67), mesenchymal-epithelial transition (E-cadherin and Vimentin), and decidualization markers (PRLR and IGFBP-1) in mouse decidual tissue on the eighth day of pregnancy were observed. RESULTS The decidual tissue structure of RSA patients was abnormal. Immunohistochemical analysis revealed significantly reduced positive expression of SGK1, ENaC-a, ERβ, and PR proteins in the decidual tissue of RSA patients (P < 0.001). Western blot and RT-PCR analyses demonstrated significantly decreased protein and mRNA expression of SGK1, ENaC-a, ERβ, and PR in the decidual tissue of RSA patients (all P < 0.05). Additionally, protein expression of PRLR and IGFBP-1 was significantly reduced (both P < 0.001). The RSA mouse model exhibited a significant increase in embryo loss rate and decreased litter size (both P < 0.001). Treatment with DQYT and JWSTW rescued the embryo loss rate and litter size to varying extents (all P < 0.05). The protein or mRNA expression levels of SGK1, ENaC-a, ERβ, PR, Ki67, E-cadherin, vimentin, PRLR, and IGFBP-1 in RSA mice were improved to different degrees after treatment with DQYT and JWSTW (all P < 0.05). CONCLUSIONS Abnormal SGK1/ENaC signaling pathway regulation is closely associated with early endometrial damage in RSA patients. JWSTW promotes endometrial proliferation and mesenchymal-epithelial transition through the SGK1/ENaC signaling pathway, improving endometrial shedding. Consequently, JWSTW is a potential treatment for RSA.
Collapse
Affiliation(s)
- Xiaoqian Di
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Institute of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050091, China.
| | - Zibo Duan
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Institute of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050091, China.
| | - Yucong Ma
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Institute of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050091, China.
| | - Xiaodan Song
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Institute of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050091, China.
| | - Yanzhi Hao
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Institute of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050091, China.
| | - Guolei Li
- Hebei Hospital of Traditional Chinese Medicine, Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050011, China.
| | - Zhanwang Tan
- Hebei Medical University, Shijiazhuang, Hebei, 050011, China.
| | - Yingying Lou
- Hebei Hospital of Traditional Chinese Medicine, Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050011, China.
| | - Xiaohua Lin
- Hebei Hospital of Traditional Chinese Medicine, Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050011, China.
| |
Collapse
|
8
|
Toudic C, Barbeau B. Isolation of Human Villous Cytotrophoblastic Cells from Term Placenta for Transfection. Methods Mol Biol 2024; 2728:13-24. [PMID: 38019388 DOI: 10.1007/978-1-0716-3495-0_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2023]
Abstract
Isolation of villous cytotrophoblastic cells (vCTBs) from placental tissue provides the possibility of studying differentiation of these cells as well as their function. We describe herein a protocol, in which term placentas are used to isolate a population of vCTB in high quantities. These cells are subsequently used to transfect siRNA through microporation and thereby to achieve efficient silencing of targeted genes.
Collapse
Affiliation(s)
- Caroline Toudic
- Département des sciences biologiques, Centre d'excellence en recherche sur les maladies orphelines-Fondation Courtois, Université du Québec à Montréal, Montreal, QC, Canada
| | - Benoit Barbeau
- Département des sciences biologiques, Centre d'excellence en recherche sur les maladies orphelines-Fondation Courtois, Université du Québec à Montréal, Montreal, QC, Canada.
- Réseau intersectoriel de recherche en santé de l'Université du Québec, Montreal, QC, Canada.
| |
Collapse
|
9
|
Di X, Hao Y, Duan Z, Ma Y, Cao Y, Tan Z, Song C, Lin X. Activation of SGK1/ENaC Signaling Pathway Improves the Level of Decidualization in Unexplained Recurrent Spontaneous Abortion. Reprod Sci 2023; 30:3273-3284. [PMID: 37280474 PMCID: PMC10643273 DOI: 10.1007/s43032-023-01273-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/11/2023] [Indexed: 06/08/2023]
Abstract
Recurrent spontaneous abortion (RSA) is one of the most common complications during pregnancy and seriously affects women's physical and mental health. About 50% of RSA cases are of unknown etiology. Our previous study found that the decidual tissue of patients with unexplained recurrent spontaneous abortion (URSA) had low expression levels of serum and glucocorticoid-induced protein kinase (SGK) 1. Endometrial decidualization is a key link in the early stage of pregnancy and is crucial to the development and maintenance of pregnancy. Decidualization is the proliferation and differentiation of endometrial stromal cells into deciduals, which involves a complex physiological process such as ovarian steroid hormones (estrogen, progesterone, prolactin, etc.), growth factors, and intercellular signaling. The binding of estrogen and its receptor stimulates the synthesis of endometrial deciduating markers prolactin (PRL) and insulin-like growth factor binding protein 1 (IGFBP-1), which mediates the occurrence of decidualization. Among them, SGK1/ENaC is a signaling pathway closely related to decidualization. The purpose of this study was to further investigate the expression of SGK1 and decidualization-related molecules in the decidual tissue of URSA patients and to explore the potential mechanism of SGK1's protective effect in URSA patients and in mouse models. Decidual tissue samples from 30 URSA patients and 30 women who actively terminated pregnancy were collected, and a URSA mouse model was established and treated with dydrogesterone. Expression levels of SGK1 and its signaling pathway-related proteins (p-Nedd4-2, 14-3-3 protein and ENaC-a), estrogen and progesterone receptors (ERβ, PR), and decidualization markers (PRLR, IGFBP-1) were assessed. Our study found that SGK1, p-Nedd4-2, 14-3-3 proteins, and ENaC-a expression levels were reduced in the decidual tissue, the SGK1/ENaC signaling pathway was inhibited, and the expression levels of the decidualization markers PRLR and IGFBP-1 were downregulated in the URSA group compared with the controls. Additionally, the concentrations of E2, P, and PRL in the serum of mice were decreased in the URSA group compared with the controls. However, SGK1/ENaC pathway-related proteins, estrogen and progesterone and their receptors, and decidualization-related molecules were upregulated by dydrogesterone. These data suggest that estrogen and progesterone can induce decidualization by activating the SGK1/ENaC signaling pathway; disruption of this pathway can lead to the development of URSA. Dydrogesterone can increase the expression level of SGK1 protein in decidual tissue.
Collapse
Affiliation(s)
- Xiaoqian Di
- Hebei University of Chinese Medicine, Shijiazhuang, 050091, Hebei, China
- Collaborative Innovation Center of Integrated Chinese and Western Medicine On Reproductive Disease, Hebei University of Chinese Medicine, Shijiazhuang, 050091, Hebei, China
- Hebei Key Laboratory of Integrative Medicine On Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, 050091, Hebei, China
| | - Yanzhi Hao
- Hebei University of Chinese Medicine, Shijiazhuang, 050091, Hebei, China
- Collaborative Innovation Center of Integrated Chinese and Western Medicine On Reproductive Disease, Hebei University of Chinese Medicine, Shijiazhuang, 050091, Hebei, China
- Hebei Key Laboratory of Integrative Medicine On Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, 050091, Hebei, China
| | - Zibo Duan
- Hebei University of Chinese Medicine, Shijiazhuang, 050091, Hebei, China
- Collaborative Innovation Center of Integrated Chinese and Western Medicine On Reproductive Disease, Hebei University of Chinese Medicine, Shijiazhuang, 050091, Hebei, China
- Hebei Key Laboratory of Integrative Medicine On Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, 050091, Hebei, China
| | - Yucong Ma
- Hebei University of Chinese Medicine, Shijiazhuang, 050091, Hebei, China
- Collaborative Innovation Center of Integrated Chinese and Western Medicine On Reproductive Disease, Hebei University of Chinese Medicine, Shijiazhuang, 050091, Hebei, China
- Hebei Key Laboratory of Integrative Medicine On Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, 050091, Hebei, China
| | - Ying Cao
- North China University of Science and Technology, Shijiazhuang, 050011, Hebei, China
| | - Zhanwang Tan
- Hebei Medical University, Shijiazhuang, 050011, Hebei, China
| | - Cuimiao Song
- Hebei University of Chinese Medicine, Shijiazhuang, 050091, Hebei, China
- Collaborative Innovation Center of Integrated Chinese and Western Medicine On Reproductive Disease, Hebei University of Chinese Medicine, Shijiazhuang, 050091, Hebei, China
- Hebei Key Laboratory of Integrative Medicine On Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, 050091, Hebei, China
| | - Xiaohua Lin
- Collaborative Innovation Center of Integrated Chinese and Western Medicine On Reproductive Disease, Hebei University of Chinese Medicine, Shijiazhuang, 050091, Hebei, China.
- Hebei Key Laboratory of Integrative Medicine On Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, 050091, Hebei, China.
- Hebei Hospital of Traditional Chinese Medicine: Affiliated Hospital of Hebei University of Chinese Medicine, Shijiazhuang, 050011, Hebei, China.
| |
Collapse
|
10
|
de Jesus Nascimento AE, Santos LC, Santos BR, Santos EO, Cunha MCDSG, Snoeck PPDN, de Lavor MSL, Silva JF. Spatial and temporal expression profile of sex steroid receptors and antioxidant enzymes in the maternal-fetal interface of domestic cats. Theriogenology 2023; 210:234-243. [PMID: 37542738 DOI: 10.1016/j.theriogenology.2023.08.001] [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: 06/08/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/07/2023]
Abstract
Sex steroids and antioxidant enzymes modulate uterine and placental physiology. Failures in the expression, signaling, and/or secretion of these mediators are associated with female infertility and gestational problems. However, there is no data on the expression profile of receptors for sex steroids and antioxidant enzymes in the maternal-fetal interface of domestic cats. Uterus and placenta samples from non-pregnant diestrus cats and cats in mid- and late pregnancy were used to analyze the protein and gene expression of the receptors for estrogen alpha (ERα), progesterone (PR), and androgen (AR) and the antioxidant enzymes superoxide dismutase 1 (SOD1), catalase, and glutathione peroxidase 1 (GPX1) by immunohistochemistry and qPCR. Higher uterine expression of ERα, Pr, and Sod1 was observed in the pregnant cats, especially in mid-pregnancy, compared to non-pregnant diestrus cats, as well as reduced endometrial catalase immunostaining. In the placenta, the mRNA expression of Erα, Pr, Ar, and Gpx1 was higher in late pregnancy in relation to mid-pregnancy. Moreover, weak or no placental expression was observed for catalase in mid- and late pregnancy, while strong immunostaining was observed for AR in trophoblasts and decidual cells in mid-pregnancy. The findings of this study demonstrated that pregnancy in female cats increases the uterine expression of sex steroid receptors and antioxidant enzymes, and that the placental expression of these mediators varies according to gestational age.
Collapse
Affiliation(s)
- Acácia Eduarda de Jesus Nascimento
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Luciano Cardoso Santos
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Bianca Reis Santos
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Emilly Oliveira Santos
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Maria Clara da Silva Galrão Cunha
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Paola Pereira das Neves Snoeck
- Hospital Veterinário, Departamento de Ciências Agrarias e Ambientais, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Mário Sérgio Lima de Lavor
- Hospital Veterinário, Departamento de Ciências Agrarias e Ambientais, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Juneo Freitas Silva
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil.
| |
Collapse
|
11
|
Li Z, Li T, Yates ME, Wu Y, Ferber A, Chen L, Brown DD, Carroll JS, Sikora MJ, Tseng GC, Oesterreich S, Lee AV. The EstroGene Database Reveals Diverse Temporal, Context-Dependent, and Bidirectional Estrogen Receptor Regulomes in Breast Cancer. Cancer Res 2023; 83:2656-2674. [PMID: 37272757 PMCID: PMC10527051 DOI: 10.1158/0008-5472.can-23-0539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/21/2023] [Accepted: 06/01/2023] [Indexed: 06/06/2023]
Abstract
As one of the most successful cancer therapeutic targets, estrogen receptor-α (ER/ESR1) has been extensively studied over the past few decades. Sequencing technological advances have enabled genome-wide analysis of ER action. However, comparison of individual studies is limited by different experimental designs, and few meta-analyses are available. Here, we established the EstroGene database through unified processing of data from 246 experiments including 136 transcriptomic, cistromic, and epigenetic datasets focusing on estradiol (E2)-triggered ER activation across 19 breast cancer cell lines. A user-friendly browser (https://estrogene.org/) was generated for multiomic data visualization involving gene inquiry under user-defined experimental conditions and statistical thresholds. Notably, annotation of metadata associated with public datasets revealed a considerable lack of experimental details. Comparison of independent RNA-seq or ER ChIP-seq data with the same design showed large variability and only strong effects could be consistently detected. Temporal estrogen response metasignatures were defined, and the association of E2 response rate with temporal transcriptional factors, chromatin accessibility, and heterogeneity of ER expression was evaluated. Unexpectedly, harmonizing 146 E2-induced transcriptomic datasets uncovered a subset of genes harboring bidirectional E2 regulation, which was linked to unique transcriptional factors and highly associated with immune surveillance in the clinical setting. Furthermore, the context dependent E2 response programs were characterized in MCF7 and T47D cell lines, the two most frequently used models in the EstroGene database. Collectively, the EstroGene database provides an informative and practical resource to the cancer research community to uniformly evaluate key reproducible features of ER regulomes and unravels modes of ER signaling. SIGNIFICANCE A resource database integrating 246 publicly available ER profiling datasets facilitates meta-analyses and identifies estrogen response temporal signatures, a bidirectional program, and model-specific biases.
Collapse
Affiliation(s)
- Zheqi Li
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh PA, USA
- Women’s Cancer Research Center, Magee Women’s Research Institute, UPMC Hillman Cancer Center, Pittsburgh PA, USA
| | - Tianqin Li
- School of Computer Science, Carnegie Mellon University, Pittsburgh PA, USA
| | - Megan E. Yates
- Women’s Cancer Research Center, Magee Women’s Research Institute, UPMC Hillman Cancer Center, Pittsburgh PA, USA
- Integrative Systems Biology Program, University of Pittsburgh, Pittsburgh, PA, USA
- Medical Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yang Wu
- Women’s Cancer Research Center, Magee Women’s Research Institute, UPMC Hillman Cancer Center, Pittsburgh PA, USA
- School of Medicine, Tsinghua University, Beijing, China
| | - Amanda Ferber
- Women’s Cancer Research Center, Magee Women’s Research Institute, UPMC Hillman Cancer Center, Pittsburgh PA, USA
| | - Lyuqin Chen
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh PA, USA
- Women’s Cancer Research Center, Magee Women’s Research Institute, UPMC Hillman Cancer Center, Pittsburgh PA, USA
| | - Daniel D. Brown
- Women’s Cancer Research Center, Magee Women’s Research Institute, UPMC Hillman Cancer Center, Pittsburgh PA, USA
- Institute for Precision Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jason S. Carroll
- Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Matthew J. Sikora
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - George C. Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh PA, USA
| | - Steffi Oesterreich
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh PA, USA
- Women’s Cancer Research Center, Magee Women’s Research Institute, UPMC Hillman Cancer Center, Pittsburgh PA, USA
- Integrative Systems Biology Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Adrian V. Lee
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh PA, USA
- Women’s Cancer Research Center, Magee Women’s Research Institute, UPMC Hillman Cancer Center, Pittsburgh PA, USA
- Integrative Systems Biology Program, University of Pittsburgh, Pittsburgh, PA, USA
- Institute for Precision Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
12
|
Lou Y, Fu Z, Tian Y, Hu M, Wang Q, Zhou Y, Wang N, Zhang Q, Jin F. Estrogen-sensitive activation of SGK1 induces M2 macrophages with anti-inflammatory properties and a Th2 response at the maternal-fetal interface. Reprod Biol Endocrinol 2023; 21:50. [PMID: 37226177 DOI: 10.1186/s12958-023-01102-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 05/15/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND Decidual macrophages participate in immune regulation at the maternal-fetal interface. Abnormal M1/M2 polarization of decidual macrophages might predispose immune maladaptation in recurrent pregnancy loss (RPL). However, the mechanism of decidual macrophage polarization is unclear. We explored the role of Estradiol (E2)-sensitive serum-glucocorticoid regulated kinase (SGK) 1 in promoting macrophage polarization and suppressing inflammation at the maternal-fetal interface. METHODS We assessed serum levels of E2 and progesterone during first trimester of pregnancy in women with or without threatened miscarriages (ended in live birth, n = 448; or early miscarriages, n = 68). For detection of SGK1 in decidual macrophages, we performed immunofluorescence labeling and western blot analysis applying decidual samples from RPL (n = 93) and early normal pregnancy (n = 66). Human monocytic THP-1 cells were differentiated into macrophages and treated with Toll-like receptor (TLR) 4 ligand lipopolysaccharide (LPS), E2, inhibitors or siRNA for in vitro analysis. Flow cytometry analysis were conducted to detect macrophages polarization. We also applied ovariectomized (OVX) mice with hormones exploring the mechanisms underlying the regulation of SGK1 activation by E2 in the decidual macrophages in vivo. RESULTS SGK1 expression down regulation in the decidual macrophages of RPL was consistent with the lower concentration and slower increment of serum E2 from 4 to 12 weeks of gestation seen in these compromised pregnancies. LPS reduced SGK1 activities, but induced the pro-inflammatory M1 phenotype of THP-1 monocyte-derived macrophages and T helper (Th) 1 cytokines that favored pregnancy loss. E2 pretreatment promoted SGK1 activation in the decidual macrophages of OVX mice in vivo. E2 pretreatment amplified SGK1 activation in TLR4-stimulated THP-1 macrophages in vitro through the estrogen receptor beta (ERβ) and PI3K pathway. E2-sensitive activation of SGK1 increased M2 macrophages and Th2 immune responses, which were beneficial to successful pregnancy, by inducing ARG1 and IRF4 transcription, which are implicated in normal pregnancy. The experiments on OVX mice have shown that pharmacological inhibition of E2 promoted nuclear translocation of NF-κB in the decidual macrophages. Further more, pharmacological inhibition or knockdown of SGK1 in TLR4-stimulated THP-1 macrophages activated NF-κB by promoting its nuclear translocation, leading to increased secretion of pro-inflammatory cytokines involved in pregnancy loss. CONCLUSION Our findings highlighted the immunomodulatory roles of E2-activated SGK1 in Th2 immune responses by priming anti-inflammatory M2 macrophages at the maternal-fetal interface, resulting in a balanced immune microenvironment during pregnancy. Our results suggest new perspectives on future preventative strategies for RPL.
Collapse
Affiliation(s)
- Yiyun Lou
- Department of Gynaecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310007, China.
| | - Zhujing Fu
- Department of Gynaecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310007, China
- Medical Department, Jinhua Municipal Central Hospital, Jinhua, 321000, China
| | - Ye Tian
- Medical School, Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Yangtze River Delta Center for Drug Evaluation and Inspection of National Medical Products Administration, Shanghai, 201210, China
| | - Minhao Hu
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Qijing Wang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Yuanyuan Zhou
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Ning Wang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Qin Zhang
- Department of Gynaecology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310007, China
| | - Fan Jin
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
- Key Laboratory of Reproductive Genetics, Women's Reproductive Healthy Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University, Hangzhou, 310006, China
| |
Collapse
|
13
|
Rusidzé M, Gargaros A, Fébrissy C, Dubucs C, Weyl A, Ousselin J, Aziza J, Arnal JF, Lenfant F. Estrogen Actions in Placental Vascular Morphogenesis and Spiral Artery Remodeling: A Comparative View between Humans and Mice. Cells 2023; 12:cells12040620. [PMID: 36831287 PMCID: PMC9954071 DOI: 10.3390/cells12040620] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/17/2023] Open
Abstract
Estrogens, mainly 17β-estradiol (E2), play a critical role in reproductive organogenesis, ovulation, and fertility via estrogen receptors. E2 is also a well-known regulator of utero-placental vascular development and blood-flow dynamics throughout gestation. Mouse and human placentas possess strikingly different morphological configurations that confer important reproductive advantages. However, the functional interplay between fetal and maternal vasculature remains similar in both species. In this review, we briefly describe the structural and functional characteristics, as well as the development, of mouse and human placentas. In addition, we summarize the current knowledge regarding estrogen actions during utero-placental vascular morphogenesis, which includes uterine angiogenesis, the control of trophoblast behavior, spiral artery remodeling, and hemodynamic adaptation throughout pregnancy, in both mice and humans. Finally, the estrogens that are present in abnormal placentation are also mentioned. Overall, this review highlights the importance of the actions of estrogens in the physiology and pathophysiology of placental vascular development.
Collapse
Affiliation(s)
- Mariam Rusidzé
- Institute of Metabolic and Cardiovascular Diseases (I2MC), INSERM U1297, University of Toulouse III-Paul Sabatier (UPS), CHU, 31432 Toulouse, France
- Department of Pathology, Cancer University Institute of Toulouse Oncopole-IUCT, 31100 Toulouse, France
| | - Adrien Gargaros
- Institute of Metabolic and Cardiovascular Diseases (I2MC), INSERM U1297, University of Toulouse III-Paul Sabatier (UPS), CHU, 31432 Toulouse, France
| | - Chanaëlle Fébrissy
- Institute of Metabolic and Cardiovascular Diseases (I2MC), INSERM U1297, University of Toulouse III-Paul Sabatier (UPS), CHU, 31432 Toulouse, France
| | - Charlotte Dubucs
- Department of Pathology, Cancer University Institute of Toulouse Oncopole-IUCT, 31100 Toulouse, France
| | - Ariane Weyl
- Institute of Metabolic and Cardiovascular Diseases (I2MC), INSERM U1297, University of Toulouse III-Paul Sabatier (UPS), CHU, 31432 Toulouse, France
- Department of Pathology, Cancer University Institute of Toulouse Oncopole-IUCT, 31100 Toulouse, France
| | - Jessie Ousselin
- Department of Pathology, Cancer University Institute of Toulouse Oncopole-IUCT, 31100 Toulouse, France
| | - Jacqueline Aziza
- Department of Pathology, Cancer University Institute of Toulouse Oncopole-IUCT, 31100 Toulouse, France
| | - Jean-François Arnal
- Institute of Metabolic and Cardiovascular Diseases (I2MC), INSERM U1297, University of Toulouse III-Paul Sabatier (UPS), CHU, 31432 Toulouse, France
| | - Françoise Lenfant
- Institute of Metabolic and Cardiovascular Diseases (I2MC), INSERM U1297, University of Toulouse III-Paul Sabatier (UPS), CHU, 31432 Toulouse, France
- Correspondence:
| |
Collapse
|
14
|
Li Z, Li T, Yates ME, Wu Y, Ferber A, Chen L, Brown DD, Carroll JS, Sikora MJ, Tseng GC, Oesterreich S, Lee AV. EstroGene database reveals diverse temporal, context-dependent and directional estrogen receptor regulomes in breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.30.526388. [PMID: 36778377 PMCID: PMC9915613 DOI: 10.1101/2023.01.30.526388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
As one of the most successful cancer therapeutic targets, estrogen receptor-α (ER/ESR1) has been extensively studied in decade-long. Sequencing technological advances have enabled genome-wide analysis of ER action. However, reproducibility is limited by different experimental design. Here, we established the EstroGene database through centralizing 246 experiments from 136 transcriptomic, cistromic and epigenetic datasets focusing on estradiol-treated ER activation across 19 breast cancer cell lines. We generated a user-friendly browser ( https://estrogene.org/ ) for data visualization and gene inquiry under user-defined experimental conditions and statistical thresholds. Notably, documentation-based meta-analysis revealed a considerable lack of experimental details. Comparison of independent RNA-seq or ER ChIP-seq data with the same design showed large variability and only strong effects could be consistently detected. We defined temporal estrogen response metasignatures and showed the association with specific transcriptional factors, chromatin accessibility and ER heterogeneity. Unexpectedly, harmonizing 146 transcriptomic analyses uncovered a subset of E2-bidirectionally regulated genes, which linked to immune surveillance in the clinical setting. Furthermore, we defined context dependent E2 response programs in MCF7 and T47D cell lines, the two most frequently used models in the field. Collectively, the EstroGene database provides an informative resource to the cancer research community and reveals a diverse mode of ER signaling.
Collapse
Affiliation(s)
- Zheqi Li
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh PA, USA
- Women’s Cancer Research Center, Magee Women’s Research Institute, UPMC Hillman Cancer Center, Pittsburgh PA, USA
| | - Tianqin Li
- School of Computer Science, Carnegie Mellon University, Pittsburgh PA, USA
| | - Megan E. Yates
- Women’s Cancer Research Center, Magee Women’s Research Institute, UPMC Hillman Cancer Center, Pittsburgh PA, USA
- Integrative Systems Biology Program, University of Pittsburgh, Pittsburgh, PA, USA
- Medical Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yang Wu
- Women’s Cancer Research Center, Magee Women’s Research Institute, UPMC Hillman Cancer Center, Pittsburgh PA, USA
- School of Medicine, Tsinghua University, Beijing, China
| | - Amanda Ferber
- Women’s Cancer Research Center, Magee Women’s Research Institute, UPMC Hillman Cancer Center, Pittsburgh PA, USA
| | - Lyuqin Chen
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh PA, USA
- Women’s Cancer Research Center, Magee Women’s Research Institute, UPMC Hillman Cancer Center, Pittsburgh PA, USA
| | - Daniel D. Brown
- Women’s Cancer Research Center, Magee Women’s Research Institute, UPMC Hillman Cancer Center, Pittsburgh PA, USA
- Institute for Precision Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jason S. Carroll
- Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Matthew J. Sikora
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - George C. Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh PA, USA
| | - Steffi Oesterreich
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh PA, USA
- Women’s Cancer Research Center, Magee Women’s Research Institute, UPMC Hillman Cancer Center, Pittsburgh PA, USA
- Integrative Systems Biology Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Adrian V. Lee
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh PA, USA
- Women’s Cancer Research Center, Magee Women’s Research Institute, UPMC Hillman Cancer Center, Pittsburgh PA, USA
- Integrative Systems Biology Program, University of Pittsburgh, Pittsburgh, PA, USA
- Institute for Precision Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
15
|
Zhu Y, Wu F, Hu J, Xu Y, Zhang J, Li Y, Lin Y, Liu X. LDHA deficiency inhibits trophoblast proliferation via the PI3K/AKT/FOXO1/CyclinD1 signaling pathway in unexplained recurrent spontaneous abortion. FASEB J 2023; 37:e22744. [PMID: 36583693 DOI: 10.1096/fj.202201219rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/05/2022] [Accepted: 12/16/2022] [Indexed: 12/31/2022]
Abstract
Dysregulated trophoblast proliferation, invasion, and apoptosis may cause several pregnancy-associated complications, such as unexplained recurrent spontaneous abortion (URSA). Recent studies have shown that metabolic abnormalities, including glycolysis inhibition, may dysregulate trophoblast function, leading to URSA. However, the underlying mechanisms remain unclear. Herein, we found that lactate dehydrogenase A (LDHA), a key enzyme in glycolysis, was significantly reduced in the placental villus of URSA patients. The human trophoblast cell line HTR-8/SVneo was used to investigate the possible LDHA-mediated regulation of trophoblast function. LDHA knockdown in HTR-8/SVneo cells induced G0/G1 phase arrest and increased apoptosis, whereas LDHA overexpression reversed these effects. Next, RNA sequencing combined with Kyoto Encyclopedia of Genes and Genomes analysis demonstrated that the PI3K/AKT signaling pathway is potentially affected by downstream genes of LDHA. Especially, we found that LDHA knockdown decreased the phosphorylation levels of PI3K, AKT, and FOXO1, resulting in a significant downregulation of CyclinD1. In addition, treatment with an AKT inhibitor or FOXO1 inhibitor also verified that the PI3K/AKT/FOXO1 signaling pathway influenced the gene expression of CyclinD1 in trophoblast. Moreover, p-AKT expression correlated positively with LDHA expression in syncytiotrophoblasts and extravillous trophoblasts in first-trimester villus. Collectively, this study revealed a new regulatory pathway for LDHA/PI3K/AKT/FOXO1/CyclinD1 in the trophoblast cell cycle and proliferation.
Collapse
Affiliation(s)
- Yueyue Zhu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China.,Institute of Birth Defects and Rare Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fan Wu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianing Hu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China.,Institute of Birth Defects and Rare Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yichi Xu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China.,Institute of Birth Defects and Rare Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jinwen Zhang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China.,Institute of Birth Defects and Rare Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Li
- Center for Reproductive Medicine, Shandong University, Jinan, China
| | - Yi Lin
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiaorui Liu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China.,Institute of Birth Defects and Rare Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
16
|
Rusidzé M, Faure MC, Sicard P, Raymond-Letron I, Giton F, Vessieres E, Prevot V, Henrion D, Arnal JF, Cornil CA, Lenfant F. Loss of function of the maternal membrane oestrogen receptor ERα alters expansion of trophoblast cells and impacts mouse fertility. Development 2022; 149:dev200683. [PMID: 36239412 PMCID: PMC9720743 DOI: 10.1242/dev.200683] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 08/31/2022] [Indexed: 03/31/2024]
Abstract
The binding of 17β-oestradiol to oestrogen receptor alpha (ERα) plays a crucial role in the control of reproduction, acting through both nuclear and membrane-initiated signalling. To study the physiological role of membrane ERα in the reproductive system, we used the C451A-ERα mouse model with selective loss of function of membrane ERα. Despite C451A-ERα mice being described as sterile, daily weighing and ultrasound imaging revealed that homozygous females do become pregnant, allowing the investigation of the role of ERα during pregnancy for the first time. All neonatal deaths of the mutant offspring mice resulted from delayed parturition associated with failure in pre-term progesterone withdrawal. Moreover, pregnant C451A-ERα females exhibited partial intrauterine embryo arrest at about E9.5. The observed embryonic lethality resulted from altered expansion of Tpbpa-positive spiral artery-associated trophoblast giant cells into the utero-placental unit, which is associated with an imbalance in expression of angiogenic factors. Together, these processes control the trophoblast-mediated spiral arterial remodelling. Hence, loss of membrane ERα within maternal tissues clearly alters the activity of invasive trophoblast cells during placentogenesis. This previously unreported function of membrane ERα could open new avenues towards a better understanding of human pregnancy-associated pathologies.
Collapse
Affiliation(s)
- Mariam Rusidzé
- Institute of Metabolic and Cardiovascular Diseases (I2MC) Equipe 4, Inserm U1297-UPS, CHU, Toulouse 31432, France
| | | | - Pierre Sicard
- IPAM, BioCampus Montpellier, CNRS, INSERM, University of Montpellier, Montpellier 34295, France
| | - Isabelle Raymond-Letron
- Institut Restore, Université de Toulouse, CNRS U-5070, EFS, ENVT, Inserm U1031, Toulouse 31076, France
| | - Frank Giton
- APHP H.Mondor - IMRB - INSERM U955, Créteil 94010, France
| | - Emilie Vessieres
- Angers University, MITOVASC, CarMe team, CNRS UMR 6015, INSERM U1083, Angers 49055, France
| | - Vincent Prevot
- University of Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR-S 1172, FHU 1000 Days for Health, Lille 59000, France
| | - Daniel Henrion
- Angers University, MITOVASC, CarMe team, CNRS UMR 6015, INSERM U1083, Angers 49055, France
| | | | | | - Françoise Lenfant
- Institute of Metabolic and Cardiovascular Diseases (I2MC) Equipe 4, Inserm U1297-UPS, CHU, Toulouse 31432, France
| |
Collapse
|
17
|
Liu M, Lian B, Lan Z, Sun H, Zhao Y, Sun T, Meng Z, Zhao C, Zhang J. Transcriptomic Profile Identifies Hippocampal Sgk1 as the Key Mediator of Ovarian Estrogenic Regulation on Spatial Learning and Memory and Aβ Accumulation. Neurochem Res 2022; 47:3369-3384. [PMID: 35915371 DOI: 10.1007/s11064-022-03690-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/14/2022] [Accepted: 07/12/2022] [Indexed: 11/28/2022]
Abstract
Previous studies have shown that ovarian estrogens are involved in the occurrence and pathology of Alzheimer's disease (AD) through regulation on hippocampal synaptic plasticity and spatial memory; however, the underlying mechanisms have not yet been elucidated at the genomic scale. In this study, we established the postmenopausal estrogen-deficient model by ovariectomy (OVX). Then, we used high-throughput Affymetrix Clariom transcriptomics and found 143 differentially expressed genes in the hippocampus of OVX mice with the absolute fold change ≥ 1.5 and P < 0.05. GO analysis showed that the highest enrichment was seen in long-term memory. Combined with the response to steroid hormone enrichment and GeneMANIA network prediction, the serum and glucocorticoid-regulated kinase 1 gene (Sgk1) was found to be the most potent candidate for ovarian estrogenic regulation. Sgk1 overexpression viral vectors (oSgk1) were then constructed and injected into the hippocampus of OVX mice. Morris water maze test revealed that the impaired spatial learning and memory induced by OVX was rescued by Sgk1 overexpression. Additionally, the altered expression of synaptic proteins and actin remodeling proteins and changes in CA1 spine density and synapse density induced by OVX were also significantly reversed by oSgk1. Moreover, the OVX-induced increase in Aβ-producing BACE1 and Aβ and the decrease in insulin degrading enzyme were significantly reversed by oSgk1. The above results show that multiple pathways and genes are involved in ovarian estrogenic regulation of the function of the hippocampus, among which Sgk1 may be a novel potent target against estrogen-sensitive hippocampal dysfunctions, such as Aβ-initiated AD.
Collapse
Affiliation(s)
- Mengying Liu
- The 305 Hospital of PLA, Beijing, 100017, China.,Department of Neurobiology, Army Medical University, Chongqing, 400038, China
| | - Biyao Lian
- Department of Pediatrics, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China.,Department of Human Anatomy and Tissue Embryology, Ningxia Medical University, Yinchuan, 750004, China
| | - Zhen Lan
- Department of Neurobiology, Army Medical University, Chongqing, 400038, China
| | - Huan Sun
- Department of Neurobiology, Army Medical University, Chongqing, 400038, China.,Center for Brain Science, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yangang Zhao
- Department of Neurology, Hainan Hospital of PLA General Hospital, Sanya, 572013, China
| | - Tao Sun
- Department of Neurobiology, Army Medical University, Chongqing, 400038, China
| | - Zhaoyou Meng
- Department of Neurobiology, Army Medical University, Chongqing, 400038, China
| | - Chengjun Zhao
- Department of Human Anatomy and Tissue Embryology, Ningxia Medical University, Yinchuan, 750004, China. .,Medical Sci-Tech Research Center, Ningxia Medical University, Yinchuan, 750004, China.
| | - Jiqiang Zhang
- Department of Neurobiology, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
18
|
Jing MY, Xie LD, Chen X, Zhou Y, Jin MM, He WH, Wang DM, Liu AX. Circ-CCNB1 Modulates Trophoblast Proliferation and Invasion in Spontaneous Abortion by Regulating miR-223/SIAH1 axis. Endocrinology 2022; 163:6613327. [PMID: 35731831 PMCID: PMC9290912 DOI: 10.1210/endocr/bqac093] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Indexed: 11/23/2022]
Abstract
CONTEXT Spontaneous abortion (SA) is a common disorder in early pregnancy. Circular RNAs (circRNAs) have been reported to exert important regulatory effects on trophoblast function and embryo development. OBJECTIVE The aim of this study was to explore whether and how circRNAs regulate trophoblast function in SA during early pregnancy. METHODS Cell proliferation, 5-bromo-2-deoxyuridine (BrdU) staining, Transwell, immunofluorescence, Western blot, RNA pull-down, and dual luciferase reporter assays were performed to investigate the effect of circRNA cyclin B1 (circ-CCNB1) on trophoblast function in HTR-8/SVneo and JEG-3 cells. RESULTS An in vitro study demonstrated that upregulation of circ-CCNB1 significantly inhibited trophoblast proliferation and invasion compared with the controls using HTR-8/SVneo and JEG-3 cells, respectively. Moreover, miR-223 was downregulated in the villous tissues of patients with SA and was further predicted and shown to negatively interact with circ-CCNB1, which is involved in trophoblast proliferation and invasion. Using bioinformatics tools and subsequent RNA pull-down and dual luciferase assays, we found that miR-223 directly targets seven in absentia homolog-1 (SIAH1) and that upregulation of miR-223 decreased circ-CCNB1-induced SIAH1 expression levels in HTR-8/SVneo cells. Interestingly, upregulation of circ-CCNB1 suppressed trophoblast proliferation and invasion through inhibition of CCNB1 nuclear translocation induced by SIAH1. Downregulation of SIAH1 enhanced circ-CCNB1-suppressed CCNB1 nuclear protein expression in trophoblast cells. CONCLUSION Circ-CCNB1 served as a modulator of trophoblast proliferation and invasion by sponging miR-223, thus forming a regulatory network of circ-CCNB1/miR-223/SIAH1 in modulating CCNB1 nuclear translocation, which enabled us to elucidate the molecular mechanisms involved in normal embryo implantation or in SA.
Collapse
Affiliation(s)
- Meng-yu Jing
- Department of Reproductive Endocrinology, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, PR China
| | - Lai-di Xie
- Department of Reproductive Endocrinology, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, PR China
| | - Xi Chen
- Department of Reproductive Endocrinology, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, PR China
| | - Ying Zhou
- Department of Reproductive Endocrinology, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, PR China
| | - Meng-meng Jin
- Department of Reproductive Endocrinology, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, PR China
| | - Wei-hua He
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, 310003, PR China
| | - Di-min Wang
- Correspondence: Di-min Wang, Department of Reproductive Endocrinology, Women’s Hospital, Zhejiang University School of Medicine, 1 Xue-shi Road, Hangzhou, 310006, China.
| | - Ai-xia Liu
- Correspondence: Ai-xia Liu, Department of Reproductive Endocrinology, Women’s Hospital, Zhejiang University School of Medicine, 1 Xue-shi Road, Hangzhou, 310006, China.
| |
Collapse
|
19
|
Johansen S, Traynor S, Ebstrup ML, Terp MG, Pedersen CB, Ditzel HJ, Gjerstorff MF. ZBED1 Regulates Genes Important for Multiple Biological Processes of the Placenta. Genes (Basel) 2022; 13:genes13010133. [PMID: 35052473 PMCID: PMC8775481 DOI: 10.3390/genes13010133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/04/2022] [Accepted: 01/11/2022] [Indexed: 02/05/2023] Open
Abstract
The transcription factor ZBED1 is highly expressed in trophoblast cells, but its functions in the processes of trophoblast and placental biology remain elusive. Here, we characterized the role of ZBED1 in trophoblast cell differentiation using an in vitro BeWo cell model. We demonstrate that ZBED1 is enhanced in its expression early after forskolin-induced differentiation of BeWo cells and regulates many of the genes that are differentially expressed as an effect of forskolin treatment. Specifically, genes encoding markers for the differentiation of cytotrophoblast into syncytiotrophoblast and factors essential for trophoblast cell fusion and invasion were negatively regulated by ZBED1, indicating that ZBED1 might be important for maintaining a steady pool of cytotrophoblast cells. In addition, ZBED1 affected genes involved in the regulation of trophoblast cell survival and apoptosis, in agreement with the observed increase in apoptosis upon knockdown of ZBED1 in forskolin-treated BeWo cells. In addition, genes implicated in the differentiation, recruitment, and function of innate immune cells by the placenta were affected by ZBED1, further suggesting a role for this protein in the regulation of maternal immune tolerance. In conclusion, our study implicates ZBED1 in major biological processes of placental biology.
Collapse
Affiliation(s)
- Simone Johansen
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (S.J.); (S.T.); (M.L.E.); (M.G.T.); (C.B.P.); (H.J.D.)
| | - Sofie Traynor
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (S.J.); (S.T.); (M.L.E.); (M.G.T.); (C.B.P.); (H.J.D.)
| | - Malene Laage Ebstrup
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (S.J.); (S.T.); (M.L.E.); (M.G.T.); (C.B.P.); (H.J.D.)
| | - Mikkel Green Terp
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (S.J.); (S.T.); (M.L.E.); (M.G.T.); (C.B.P.); (H.J.D.)
| | - Christina Bøg Pedersen
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (S.J.); (S.T.); (M.L.E.); (M.G.T.); (C.B.P.); (H.J.D.)
| | - Henrik Jørn Ditzel
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (S.J.); (S.T.); (M.L.E.); (M.G.T.); (C.B.P.); (H.J.D.)
- Department of Oncology, Odense University Hospital, 5230 Odense, Denmark
- Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, 5230 Odense, Denmark
| | - Morten Frier Gjerstorff
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (S.J.); (S.T.); (M.L.E.); (M.G.T.); (C.B.P.); (H.J.D.)
- Department of Oncology, Odense University Hospital, 5230 Odense, Denmark
- Academy of Geriatric Cancer Research (AgeCare), Odense University Hospital, 5230 Odense, Denmark
- Correspondence: ; Tel.: +45-2126-1563
| |
Collapse
|
20
|
Carey AZ, Blue NR, Varner MW, Page JM, Chaiyakunapruk N, Quinlan AR, Branch DW, Silver RM, Workalemahu T. A Systematic Review to Guide Future Efforts in the Determination of Genetic Causes of Pregnancy Loss. FRONTIERS IN REPRODUCTIVE HEALTH 2021; 3. [PMID: 35462723 PMCID: PMC9031276 DOI: 10.3389/frph.2021.770517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background: Pregnancy loss is the most common obstetric complication occurring in almost 30% of conceptions overall and in 12–14% of clinically recognized pregnancies. Pregnancy loss has strong genetic underpinnings, and despite this consensus, our understanding of its genetic causes remains limited. We conducted a systematic review of genetic factors in pregnancy loss to identify strategies to guide future research.Methods: To synthesize data from population-based association studies on genetics of pregnancy loss, we searched PubMed for relevant articles published between 01/01/2000-01/01/2020. We excluded review articles, case studies, studies with limited sample sizes to detect associations (N < 4), descriptive studies, commentaries, and studies with non-genetic etiologies. Studies were classified based on developmental periods in gestation to synthesize data across various developmental epochs.Results: Our search yielded 580 potential titles with 107 (18%) eligible after title/abstract review. Of these, 54 (50%) were selected for systematic review after full-text review. These studies examined either early pregnancy loss (n = 9 [17%]), pregnancy loss >20 weeks' gestation (n = 10 [18%]), recurrent pregnancy loss (n = 32 [59%]), unclassified pregnancy loss (n = 3 [4%]) as their primary outcomes. Multiple genetic pathways that are essential for embryonic/fetal survival as well as human development were identified.Conclusion: Several genetic pathways may play a role in pregnancy loss across developmental periods in gestation. Systematic evaluation of pregnancy loss across developmental epochs, utilizing whole genome sequencing in families may further elucidate causal genetic mechanisms and identify other pathways critical for embryonic/fetal survival.
Collapse
Affiliation(s)
- Andrew Z. Carey
- Department of Obstetrics & Gynecology, University of Utah Health, Salt Lake City, UT, United States
| | - Nathan R. Blue
- Department of Obstetrics & Gynecology, University of Utah Health, Salt Lake City, UT, United States
- Department of Obstetrics and Gynecology, Intermountain Healthcare, Salt Lake City, UT, United States
| | - Michael W. Varner
- Department of Obstetrics & Gynecology, University of Utah Health, Salt Lake City, UT, United States
- Department of Obstetrics and Gynecology, Intermountain Healthcare, Salt Lake City, UT, United States
| | - Jessica M. Page
- Department of Obstetrics & Gynecology, University of Utah Health, Salt Lake City, UT, United States
- Department of Obstetrics and Gynecology, Intermountain Healthcare, Salt Lake City, UT, United States
| | - Nathorn Chaiyakunapruk
- Department of Pharmacotherapy, College of Pharmacy, University of Utah, Salt Lake City, UT, United States
- School of Pharmacy, Monash University Malaysia, Subang Jaya, Malaysia
| | - Aaron R. Quinlan
- Department of Human Genetics, University of Utah, Salt Lake City, UT, United States
- Utah Center for Genetic Discovery, University of Utah, Salt Lake City, UT, United States
- Department of Biomedical Informatics, University of Utah, Salt Lake City, UT, United States
| | - D. Ware Branch
- Department of Obstetrics & Gynecology, University of Utah Health, Salt Lake City, UT, United States
- Department of Obstetrics and Gynecology, Intermountain Healthcare, Salt Lake City, UT, United States
| | - Robert M. Silver
- Department of Obstetrics & Gynecology, University of Utah Health, Salt Lake City, UT, United States
- Department of Obstetrics and Gynecology, Intermountain Healthcare, Salt Lake City, UT, United States
| | - Tsegaselassie Workalemahu
- Department of Obstetrics & Gynecology, University of Utah Health, Salt Lake City, UT, United States
- *Correspondence: Tsegaselassie Workalemahu
| |
Collapse
|
21
|
Liu A, Jin M, Xie L, Jing M, Zhou Y, Tang M, Lin T, Wang D. Loss of miR-29a impairs decidualization of endometrial stromal cells by TET3 mediated demethylation of Col1A1 promoter. iScience 2021; 24:103065. [PMID: 34568789 PMCID: PMC8449092 DOI: 10.1016/j.isci.2021.103065] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 06/28/2021] [Accepted: 08/26/2021] [Indexed: 12/01/2022] Open
Abstract
A conceptual framework for understanding abnormal endometrial decidualization, with considerable significance for the diagnosis and treatment of abnormal decidualization-related changes in non-receptive endometrium in implantation failure during early pregnancy is very important. Here, we found the expression levels of miR-29a in endometrial tissues were associated with the menstrual phases and pregnancy outcome. Inhibition of miR-29a led to decreased decidualization of endometrial stromal cells (ESCs) in vitro, whereas Tet methylcytosine dioxygenase 3 (TET3) and its potential demethylation target, the collagen type I alpha 1 chain (Col1A1), were restored. The binding capacity of TET3 to the Col1A1 promoter could be enhanced by the inhibition of miR-29a. Finally, deletion of TET3 rescued the inhibitory effect of the miR-29a antagomir on the proliferation of decidualized ESCs in vitro and embryo implantation in vivo. Thus, loss of miR-29a causes implantation failure because of the limitation of ESCs decidualization-related changes in non-receptive endometrium during early pregnancy. Loss of miR-29a inhibits decidualization of ESCs TET3 demethylates the Col1A1 promoter Loss of miR-29a enhances the binding capacity of TET3 to the Col1A1 promoter Loss of miR-29a suppresses embryo implantation during early pregnancy in mice
Collapse
Affiliation(s)
- Aixia Liu
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, 1 Xueshi Road, Hangzhou 310006, PR China.,Key Laboratory of Reproductive Genetics (Ministry of Education), Zhejiang University, Hangzhou 310006, PR China
| | - Mengmeng Jin
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, 1 Xueshi Road, Hangzhou 310006, PR China
| | - Laidi Xie
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, 1 Xueshi Road, Hangzhou 310006, PR China
| | - Mengyu Jing
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, 1 Xueshi Road, Hangzhou 310006, PR China
| | - Ying Zhou
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, 1 Xueshi Road, Hangzhou 310006, PR China
| | - Minyue Tang
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, 1 Xueshi Road, Hangzhou 310006, PR China.,Key Laboratory of Reproductive Genetics (Ministry of Education), Zhejiang University, Hangzhou 310006, PR China
| | - Tingting Lin
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, 1 Xueshi Road, Hangzhou 310006, PR China
| | - Dimin Wang
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, 1 Xueshi Road, Hangzhou 310006, PR China
| |
Collapse
|
22
|
Cañumil VA, Bogetti E, de la Cruz Borthiry FL, Ribeiro ML, Beltrame JS. Steroid hormones and first trimester vascular remodeling. VITAMINS AND HORMONES 2021; 116:363-387. [PMID: 33752825 DOI: 10.1016/bs.vh.2021.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Successful implantation and placentation require neoangiogenesis and the remodeling of the uterine spiral arteries. Progesterone and estradiol control various of the placental functions, but their role in vascular remodeling remains controversial. Therefore, this chapter aims to summarize the current knowledge regarding the role of steroid hormones in the uteroplacental vascular remodeling during the first trimester of gestation.
Collapse
Affiliation(s)
- V A Cañumil
- Center of Pharmacological and Botanical Studies (CEFyBO), School of Medicine University of Buenos Aires (UBA)-National Research Council (CONICET), Buenos Aires, Argentina
| | - E Bogetti
- Center of Pharmacological and Botanical Studies (CEFyBO), School of Medicine University of Buenos Aires (UBA)-National Research Council (CONICET), Buenos Aires, Argentina
| | - F L de la Cruz Borthiry
- Center of Pharmacological and Botanical Studies (CEFyBO), School of Medicine University of Buenos Aires (UBA)-National Research Council (CONICET), Buenos Aires, Argentina
| | - M L Ribeiro
- Center of Pharmacological and Botanical Studies (CEFyBO), School of Medicine University of Buenos Aires (UBA)-National Research Council (CONICET), Buenos Aires, Argentina
| | - J S Beltrame
- Center of Pharmacological and Botanical Studies (CEFyBO), School of Medicine University of Buenos Aires (UBA)-National Research Council (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
23
|
Liu Y, Ma H, Yao J. ERα, A Key Target for Cancer Therapy: A Review. Onco Targets Ther 2020; 13:2183-2191. [PMID: 32210584 PMCID: PMC7073439 DOI: 10.2147/ott.s236532] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 02/20/2020] [Indexed: 12/18/2022] Open
Abstract
Estrogen receptor α (ERα) is closely associated with both hormone-dependent and hormone-independent tumors, and it is also essential for the development of these cancers. The functions of ERα are bi-faceted; it can contribute to cancer progression as well as cancer inhibition. Therefore, understanding ERα is vital for the treatment of those cancers that are closely associated with its expression. Here, we will elaborate on ERα based on its structure, localization, activation, modification, and mutation. Also, we will look at co-activators of ERα, elucidate the signaling pathway activated by ERα, and identify cancers related to its activation. A comprehensive understanding of ERα could help us to find new ways to treat cancers.
Collapse
Affiliation(s)
- Yanfang Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Hong Ma
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Jing Yao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| |
Collapse
|