1
|
Cao D, Liu Y, Cheng Y, Wang J, Zhang B, Zhai Y, Zhu K, Liu Y, Shang Y, Xiao X, Chang Y, Lee YL, Yeung WSB, Huang Y, Yao Y. Time-series single-cell transcriptomic profiling of luteal-phase endometrium uncovers dynamic characteristics and its dysregulation in recurrent implantation failures. Nat Commun 2025; 16:137. [PMID: 39747825 PMCID: PMC11695634 DOI: 10.1038/s41467-024-55419-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 12/09/2024] [Indexed: 01/04/2025] Open
Abstract
Understanding human endometrial dynamics in the establishment of endometrial receptivity remains a challenge, which limits early diagnosis and treatment of endometrial-factor infertility. Here, we decode the endometrial dynamics of fertile women across the window of implantation and characterize the endometrial deficiency in women with recurrent implantation failure. A computational model capable of both temporal prediction and pattern discovery is used to analyze single-cell transcriptomic data from over 220,000 endometrial cells. The time-series atlas highlights a two-stage stromal decidualization process and a gradual transitional process of the luminal epithelial cells across the window of implantation. In addition, a time-varying gene set regulating epithelium receptivity is identified, based on which the recurrent implantation failure endometria are stratified into two classes of deficiencies. Further investigation uncovers a hyper-inflammatory microenvironment for the dysfunctional endometrial epithelial cells of recurrent implantation failure. The holistic characterization of the physiological and pathophysiological window of implantation and a computational tool trained on this temporal atlas provide a platform for future therapeutic developments.
Collapse
Affiliation(s)
- Dandan Cao
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, the University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yijun Liu
- School of Biomedical Sciences, the University of Hong Kong, Hong Kong SAR, China
- School of Artificial Intelligence, Jilin University, Jilin, China
| | - Yanfei Cheng
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, the University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Jue Wang
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, the University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Bolun Zhang
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, the University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- School of Medicine, Nankai University, Tianjin, China
| | - Yanhui Zhai
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, the University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Kongfu Zhu
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, the University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Ye Liu
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, the University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Ye Shang
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, the University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Xiao Xiao
- Genomics Institute, Geneplus-Shenzhen, Shenzhen, China
| | - Yi Chang
- School of Artificial Intelligence, Jilin University, Jilin, China
| | - Yin Lau Lee
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, the University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Obstetrics and Gynaecology, the University of Hong Kong, Hong Kong SAR, China
- Centre for Translational Stem Cell Biology, Building 17 W, The Hong Kong Science and Technology Park, Hong Kong SAR, China
| | - William Shu Biu Yeung
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, the University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
- Department of Obstetrics and Gynaecology, the University of Hong Kong, Hong Kong SAR, China.
- Centre for Translational Stem Cell Biology, Building 17 W, The Hong Kong Science and Technology Park, Hong Kong SAR, China.
| | - Yuanhua Huang
- School of Biomedical Sciences, the University of Hong Kong, Hong Kong SAR, China.
- Centre for Translational Stem Cell Biology, Building 17 W, The Hong Kong Science and Technology Park, Hong Kong SAR, China.
- Department of Statistics and Actuarial Science, the University of Hong Kong, Hong Kong SAR, China.
| | - Yuanqing Yao
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, the University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
- Department of Gynecology and Obstetrics, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
2
|
Zhang Y, Zhang Y, Xia X, Gao L, Gao C, Zhou J, Yan Z, Cui Y, Ma X, Kwak-Kim JYH, Diao F. Hyperlipidemia negatively impacts implantation by dysregulating tight junction and Claudin-3 and Claudin-4 expression in the endometrium. J Reprod Immunol 2024; 166:104326. [PMID: 39265316 DOI: 10.1016/j.jri.2024.104326] [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: 06/17/2024] [Revised: 08/07/2024] [Accepted: 09/01/2024] [Indexed: 09/14/2024]
Abstract
Clinical observational studies have suggested hyperlipidemia may disturb embryo implantation through endometrium; however, the mechanism has been unclear. With its profound implications for reproductive health, the present study aims to investigate whether hyperlipidemia affects endometrial epithelial cell tight junctions for implantation failures. By constructing hyperlipidemia mice model, the number and distribution of embryo implantation status were investigated after both natural mating and in vitro fertilization and embryo transfer (IVF-ET). Transmission electron microscopy (TEM) was used to compare the ultrastructure of tight junctions in endometrial endothelial cells. Western blot and immunofluorescence were used to explore the expression and localization of tight junction proteins, such as Claudin (CDLN)3, CLDN4, occludin (OCLN), and zonula occludens-1 (ZO1). For women with reproductive failure, mid-luteal phase endometrial tissues were collected, and gene expression of tight junction proteins was investigated using RNA sequencing and qRT-PCR. In hyperlipidemic mice, the number of embryo implantation sites significantly decreased with uneven distribution after natural mating and IVF-ET. Disrupted tight junctions were found, characterized by a decreased number of tight junctions by TEM, downregulated expressions of CDLN4, OCLN, and ZO1, and an increased expression of CLDN3 by western blot. In hyperlipidemic women with reproductive failure, the dysregulated expression of CLDN3 and CLDN4 was also present in the luteal phase endometrium. In this study, evaluation of both animal models and infertile women in vivo demonstrated that hyperlipidemia reduced female fertility, accompanied by disruption of tight junction structures and dysregulation of CLDN3 and CLDN4 expression in the endothelial cells of luteal phase endometrium.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Reproductive Medicine, Jiangsu Province Hospital, Guangzhou Road 300, Nanjing, Jiangsu 210029, China
| | - Yuexin Zhang
- Center of Clinical Laboratory Medicine of Zhongda Hospital, Southeast University, 87 Dingjiaqiao, Nanjing, Jiangsu 210009, China
| | - Xinru Xia
- Department of Reproductive Medicine, Jiangsu Province Hospital, Guangzhou Road 300, Nanjing, Jiangsu 210029, China
| | - Li Gao
- Department of Reproductive Medicine, Jiangsu Province Hospital, Guangzhou Road 300, Nanjing, Jiangsu 210029, China
| | - Chao Gao
- Department of Reproductive Medicine, Jiangsu Province Hospital, Guangzhou Road 300, Nanjing, Jiangsu 210029, China
| | - Jing Zhou
- Department of Reproductive Medicine, Jiangsu Province Hospital, Guangzhou Road 300, Nanjing, Jiangsu 210029, China
| | - Zhengjie Yan
- Department of Reproductive Medicine, Jiangsu Province Hospital, Guangzhou Road 300, Nanjing, Jiangsu 210029, China
| | - Yugui Cui
- Department of Reproductive Medicine, Jiangsu Province Hospital, Guangzhou Road 300, Nanjing, Jiangsu 210029, China
| | - Xiang Ma
- Department of Reproductive Medicine, Jiangsu Province Hospital, Guangzhou Road 300, Nanjing, Jiangsu 210029, China.
| | - Joanne Young Hee Kwak-Kim
- Reproductive Medicine and Immunology, Obstetrics and Gynecology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3471 N Green Bay Road, North Chicago, IL 60064, USA.
| | - Feiyang Diao
- Department of Reproductive Medicine, Jiangsu Province Hospital, Guangzhou Road 300, Nanjing, Jiangsu 210029, China.
| |
Collapse
|
3
|
Moura TDBD, Nunes FB, Crestani BDV, Araujo TFC, Hanauer EL, Corleta HVE, Branchini G. Preeclampsia and transport of ions and small molecules: A literature review. Placenta 2024; 156:77-91. [PMID: 39293185 DOI: 10.1016/j.placenta.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/22/2024] [Accepted: 09/12/2024] [Indexed: 09/20/2024]
Abstract
Preeclampsia (PE) is a prevalent obstetric complication affecting approximately 3-5% of pregnancies worldwide and is a major cause of maternal and perinatal morbidity and mortality. Preeclampsia is considered a disease of the endothelial system that can progress to eclampsia, characterized by seizures. Early diagnosis and appropriate management are crucial to improving maternal and fetal outcomes, as preeclampsia can lead to severe complications such as placental abruption, fetal growth restriction, and stroke. The pathophysiology of PE is complex, involving a combination of genetic, acquired, and immunological factors. A central feature of the condition is inadequate placentation and impaired uteroplacental perfusion, leading to local hypoxia, endothelial dysfunction, vasoconstriction, and immunological dysregulation. Recent evidence suggests that dysregulation of ion transporters may play a significant role in the adaptation of uterine circulation during placentation. These transporters are essential for maintaining maternal-fetal homeostasis, influencing processes such as nutrient exchange, hormone synthesis, trophoblast cell migration, and the function of smooth muscle cells in blood vessels. In preeclampsia, adverse conditions like hypoxia and oxidative stress result in the downregulation of ion, solute, and water transporters, impairing their function. This review focuses on membrane transporters involved in PE, discussing functional alterations and their physiological implications. The goal of this investigation is to enhance understanding of how dysregulation of ion and small molecule transporters contributes to the development and progression of preeclampsia, underscoring the importance of exploring these signaling pathways for potential therapeutic interventions.
Collapse
Affiliation(s)
- Thaís Duarte Borges de Moura
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), 245 Sarmento Leite St, Porto Alegre, RS, ZIP 90050170, Brazil
| | - Fernanda Bordignon Nunes
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), 245 Sarmento Leite St, Porto Alegre, RS, ZIP 90050170, Brazil; Pontifícia Universidade Católica Do Rio Grande Do Sul (PUCRS), 6681 Ipiranga Av, Porto Alegre, RS, ZIP 90619-900, Brazil
| | - Bianca Dalla Vecchia Crestani
- Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), 245 Sarmento Leite St, Porto Alegre, ZIP 90050170, Brazil
| | | | - Eduarda Luiza Hanauer
- Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), 245 Sarmento Leite St, Porto Alegre, ZIP 90050170, Brazil
| | - Helena von Eye Corleta
- Departamento de Ginecologia e Obstetrícia, Faculdade de Medicina, Universidade Federal Do Rio Grande Do Sul (UFRGS), 2400 Ramiro Barcelos St, Porto Alegre, RS, ZIP 90035-003, Brazil
| | - Gisele Branchini
- Graduate Program in Pathology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), 245 Sarmento Leite St, Porto Alegre, RS, ZIP 90050170, Brazil.
| |
Collapse
|
4
|
Davoodi Nik B, Hashemi Karoii D, Favaedi R, Ramazanali F, Jahangiri M, Movaghar B, Shahhoseini M. Differential expression of ion channel coding genes in the endometrium of women experiencing recurrent implantation failures. Sci Rep 2024; 14:19822. [PMID: 39192025 PMCID: PMC11349755 DOI: 10.1038/s41598-024-70778-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 08/21/2024] [Indexed: 08/29/2024] Open
Abstract
Our study probed the differences in ion channel gene expression in the endometrium of women with Recurrent Implantation Failure (RIF) compared to fertile women. We analyzed the relative expression of genes coding for T-type Ca2+, ENaC, CFTR, and KCNQ1 channels in endometrial samples from 20 RIF-affected and 10 control women, aged 22-35, via microarray analysis and quantitative real-time PCR. Additionally, we examined DNA methylation in the regulatory region of KCNQ1 using ChIP real-time PCR. The bioinformatics component of our research included Gene Ontology analysis, protein-protein interaction networks, and signaling pathway mapping to identify key biological processes and pathways implicated in RIF. This led to the discovery of significant alterations in the expression of ion channel genes in RIF women's endometrium, most notably an overexpression of CFTR and reduced expression of SCNN1A, SCNN1B, SCNN1G, CACNA1H, and KCNQ1. A higher DNA methylation level of KCNQ1's regulatory region was also observed in RIF patients. Gene-set enrichment analysis highlighted a significant presence of genes involved with ion transport and membrane potential regulation, particularly in sodium and calcium channel complexes, which are vital for cation movement across cell membranes. Genes were also enriched in broader ion channel and transmembrane transporter complexes, underscoring their potential extensive role in cellular ion homeostasis and signaling. These findings suggest a potential involvement of ion channels in the pathology of implantation failure, offering new insights into the mechanisms behind RIF and possible therapeutic targets.
Collapse
Affiliation(s)
- Bahar Davoodi Nik
- Department of Genetics, Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Danial Hashemi Karoii
- Department of Cell and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Raha Favaedi
- Department of Genetics, Reproductive Biomedicine Research Centre, Royan Institute for Reproductive Biomedicine, ACECR, No. 12, Hafez St., Banihashem Sq, Resalat Ave., P.O. Box: 19395-4644, Tehran, Iran
| | - Fariba Ramazanali
- Department of Endocrinology and Female Infertility, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Maryam Jahangiri
- Department of Embryology, Reproductive Biomedicine Research Centre, Royan Institute for Reproductive Biomedicine, ACECR, No. 12, Hafez St., Banihashem Sq, Resalat Ave., P.O. Box: 19395-4644, Tehran, Iran
| | - Bahar Movaghar
- Department of Embryology, Reproductive Biomedicine Research Centre, Royan Institute for Reproductive Biomedicine, ACECR, No. 12, Hafez St., Banihashem Sq, Resalat Ave., P.O. Box: 19395-4644, Tehran, Iran.
| | - Maryam Shahhoseini
- Department of Genetics, Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran.
- Department of Cell and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
- Department of Genetics, Reproductive Biomedicine Research Centre, Royan Institute for Reproductive Biomedicine, ACECR, No. 12, Hafez St., Banihashem Sq, Resalat Ave., P.O. Box: 19395-4644, Tehran, Iran.
| |
Collapse
|
5
|
Ma X, Xu R, Chen J, Wang S, Hu P, Wu Y, Que Y, Du W, Cai X, Chen H, Guo J, Li TC, Ruan YC. The epithelial Na + channel (ENaC) in ovarian granulosa cells modulates Ca 2+ mobilization and gonadotrophin signaling for estrogen homeostasis and female fertility. Cell Commun Signal 2024; 22:398. [PMID: 39143495 PMCID: PMC11323461 DOI: 10.1186/s12964-024-01778-5] [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: 05/23/2024] [Accepted: 08/06/2024] [Indexed: 08/16/2024] Open
Abstract
Ovarian granulosa cells are essential to gonadotrophin-regulated estrogen production, female cycle maintenance and fertility. The epithelial Na+ channel (ENaC) is associated with female fertility; however, whether and how it plays a role in ovarian cell function(s) remained unexplored. Here, we report patch-clamp and Na+ imaging detection of ENaC expression and channel activity in both human and mouse ovarian granulosa cells, which are promoted by pituitary gonadotrophins, follicle stimulating hormone (FSH) or luteinizing hormone (LH). Cre-recombinase- and CRISPR-Cas9-based granulosa-specific knockout of ENaC α subunit (Scnn1a) in mice resulted in failed estrogen elevation at early estrus, reduced number of corpus luteum, abnormally extended estrus phase, reduced litter size and subfertility in adult female mice. Further analysis using technologies including RNA sequencing and Ca2+ imaging revealed that pharmacological inhibition, shRNA-based knockdown or the knockout of ENaC diminished spontaneous or stimulated Ca2+ oscillations, lowered the capacity of intracellular Ca2+ stores and impaired FSH/LH-stimulated transcriptome changes for estrogen production in mouse and/or human granulosa cells. Together, these results have revealed a previously undefined role of ENaC in modulating gonadotrophin signaling in granulosa cells for estrogen homeostasis and thus female fertility.
Collapse
Affiliation(s)
- Xiyang Ma
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Ruiyao Xu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Junjiang Chen
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
- Jinan University, Guangzhou, China
| | - Shan Wang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Peijie Hu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Yong Wu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Yanting Que
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Wanting Du
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Xiaojun Cai
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Hui Chen
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Jinghui Guo
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, China
| | - Tin Chiu Li
- Department of Obstetrics and Gynaecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ye Chun Ruan
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong SAR, China.
- Shenzhen Research Institute, The Hong Kong Polytechnic University, Shenzhen, China.
| |
Collapse
|
6
|
Sadowska A, Molcan T, Wójtowicz A, Lukasik K, Pawlina-Tyszko K, Gurgul A, Ferreira-Dias G, Skarzynski DJ, Szóstek-Mioduchowska A. Bioinformatic analysis of endometrial miRNA expression profile at day 26-28 of pregnancy in the mare. Sci Rep 2024; 14:3900. [PMID: 38365979 PMCID: PMC10873421 DOI: 10.1038/s41598-024-53499-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: 12/19/2023] [Accepted: 02/01/2024] [Indexed: 02/18/2024] Open
Abstract
The establishment of the fetomaternal interface depends on precisely regulated communication between the conceptus and the uterine environment. Recent evidence suggests that microRNAs (miRNAs) may play an important role in embryo-maternal dialogue. This study aimed to determine the expression profile of endometrial miRNAs during days 26-28 of equine pregnancy. Additionally, the study aimed to predict target genes for differentially expressed miRNAs (DEmiRs) and their potential role in embryo attachment, adhesion, and implantation. Using next-generation sequencing, we identified 81 DEmiRs between equine endometrium during the pre-attachment period of pregnancy (day 26-28) and endometrium during the mid-luteal phase of the estrous cycle (day 10-12). The identified DEmiRs appear to have a significant role in regulating the expression of genes that influence cell fate and properties, as well as endometrial receptivity formation. These miRNAs include eca-miR-21, eca-miR-126-3p, eca-miR-145, eca-miR-451, eca-miR-491-5p, members of the miR-200 family, and the miRNA-17-92 cluster. The target genes predicted for the identified DEmiRs are associated with ion channel activity and sphingolipid metabolism. Furthermore, it was noted that the expression of mucin 1 and leukemia inhibitory factor, genes potentially regulated by the identified DEmiRs, was up-regulated at day 26-28 of pregnancy. This suggests that miRNAs may play a role in regulating specific genes to create a favorable uterine environment that is necessary for proper attachment, adhesion, and implantation of the embryo in mares.
Collapse
Affiliation(s)
- Agnieszka Sadowska
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Tuwima Street 10, 10-748, Olsztyn, Poland
| | - Tomasz Molcan
- Molecular Biology Laboratory, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima Street 10, 10-748, Olsztyn, Poland
| | - Anna Wójtowicz
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Tuwima Street 10, 10-748, Olsztyn, Poland
| | - Karolina Lukasik
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Tuwima Street 10, 10-748, Olsztyn, Poland
| | - Klaudia Pawlina-Tyszko
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Sarego Street 2, 31-047, Kraków, Poland
| | - Artur Gurgul
- Center for Experimental and Innovative Medicine, University of Agriculture in Krakow, Mickiewicza Street 21, 31-120, Kraków, Poland
| | - Graca Ferreira-Dias
- CIISA-Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1300-477, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477, Lisbon, Portugal
| | - Dariusz J Skarzynski
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Tuwima Street 10, 10-748, Olsztyn, Poland
| | - Anna Szóstek-Mioduchowska
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Tuwima Street 10, 10-748, Olsztyn, Poland.
| |
Collapse
|
7
|
Zahir M, Tavakoli B, Zaki-Dizaji M, Hantoushzadeh S, Majidi Zolbin M. Non-coding RNAs in Recurrent implantation failure. Clin Chim Acta 2024; 553:117731. [PMID: 38128815 DOI: 10.1016/j.cca.2023.117731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/17/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
Recurrent implantation failure (RIF), defined as the inability to achieve conception following multiple consecutive in-vitro fertilization (IVF) attempts, represents a complex and multifaceted challenge in reproductive medicine. The emerging role of non-coding RNAs in RIF etiopathogenesis has only gained prominence over the last decade, illustrating a new dimension to our understanding of the intricate network underlying RIF. Successful embryo implantation demands a harmonious synchronization between an adequately decidualized endometrium, a competent blastocyst, and effective maternal-embryonic interactions. Emerging evidence has clarified the involvement of a sophisticated network of non-coding RNAs, including microRNAs, circular RNAs, and long non-coding RNAs, in orchestrating these pivotal processes. Disconcerted expression of these molecules can disrupt the delicate equilibrium required for implantation, amplifying the risk of RIF. This comprehensive review presents an in-depth investigation of the complex role played by non-coding RNAs in the pathogenesis of RIF. Furthermore, it underscores the vast potential of non-coding RNAs as diagnostic biomarkers and therapeutic targets, with the ultimate goal of enhancing implantation success rates in IVF cycles. As ongoing research continues to unravel the intercalated web of molecular interactions, exploiting the power of non-coding RNAs may offer promising avenues for mitigating the challenges posed by RIF and improving the outcomes of assisted reproduction.
Collapse
Affiliation(s)
- Mazyar Zahir
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahareh Tavakoli
- Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Biology, Maragheh University, Maragheh, Iran
| | - Majid Zaki-Dizaji
- Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | - Sedigheh Hantoushzadeh
- Vali-E-Asr Reproductive Health Research Center, Family Health Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Masoumeh Majidi Zolbin
- Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
He D, Song Y, Xiao H, Shi S, Song H, Cui T, Ni T, Wang J, Ren X, Wei A. Ligustilide enhances pregnancy outcomes via improvement of endometrial receptivity and promotion of endometrial angiogenesis in rats. J Nat Med 2024; 78:42-52. [PMID: 37698739 DOI: 10.1007/s11418-023-01739-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 07/26/2023] [Indexed: 09/13/2023]
Abstract
Ligustilide (LIG) is the main active ingredient of Angelica sinensis (Oliv.) Diels, which could promote focal angiogenesis to exert neuroprotection. However, there was no report that verified the exact effects of LIG on endometrial angiogenesis and the pregnancy outcomes. To explore the effects of LIG on low endometrial receptivity (LER) and angiogenesis, pregnancy rats were assigned into Control (saline treatment), LER (hydroxyurea-adrenaline treatment), LIG 20 mg/kg and LIG 40 mg/kg groups. Hematoxylin and eosin (H&E) staining was performed to evaluate endometrial morphology. Quantitative real-time PCR, immunofluorescence staining, western blot and immunohistochemistry staining were employed to assess the expression of endometrial receptivity factors and angiogenesis-related gene/protein, respectively. RNA sequencing was used to analyze the effects of LIG on LER caused by Kidney deficiency and blood stasis. We found that endometrial thickness and the implanted embryo number were substantially reduced in the hydroxyurea-adrenaline-treated pregnancy rats. At the same time, the gene and protein expressions of ERα, LIF, VEGFA and CD31 in the endometrium were markedly reduced, while the expressions of MUC1, E-cadherin were increased in the LER group. Administration of LIG raised the endometrial thickness and implanted embryos, as well as reversed the expressions of these factors. Collectively, our findings revealed that LIG could facilitate embryo implantation via recovery of the endometrium receptivity and promotion of endometrial angiogenesis.
Collapse
Affiliation(s)
- Dongjie He
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, Henan, China
| | - Yanli Song
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, Henan, China
| | - Huidongzi Xiao
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, Henan, China
| | - Shaoqi Shi
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, Henan, China
| | - Hongyan Song
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, Henan, China
| | - Tianwei Cui
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, Henan, China
| | - Tingting Ni
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Jing Wang
- The First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Xingxing Ren
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, Henan, China.
| | - Aiwu Wei
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, Henan, China.
| |
Collapse
|
9
|
Abstract
Embryo implantation in humans is interstitial, meaning the entire conceptus embeds in the endometrium before the placental trophoblast invades beyond the uterine mucosa into the underlying inner myometrium. Once implanted, embryo survival pivots on the transformation of the endometrium into an anti-inflammatory placental bed, termed decidua, under homeostatic control of uterine natural killer cells. Here, we examine the evolutionary context of embryo implantation and elaborate on uterine remodelling before and after conception in humans. We also discuss the interactions between the embryo and the decidualising endometrium that regulate interstitial implantation and determine embryo fitness. Together, this Review highlights the precarious but adaptable nature of the implantation process.
Collapse
Affiliation(s)
- Joanne Muter
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV2 2DX, UK
- Tommy's National Centre for Miscarriage Research, University Hospitals Coventry & Warwickshire NHS Trust, Warwick Medical School, University of Warwick, Coventry, CV2 2DX, UK
| | - Vincent J. Lynch
- Department of Biological Sciences, University at Buffalo, Buffalo, NY 14260-4610, USA
| | - Rajiv C. McCoy
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jan J. Brosens
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV2 2DX, UK
- Tommy's National Centre for Miscarriage Research, University Hospitals Coventry & Warwickshire NHS Trust, Warwick Medical School, University of Warwick, Coventry, CV2 2DX, UK
| |
Collapse
|
10
|
Effects of maternal HF diet and absence of TRPC1 gene on mouse placental growth and fetal intrauterine growth retardation (IUGR). J Nutr Biochem 2023; 114:109162. [PMID: 36243380 DOI: 10.1016/j.jnutbio.2022.109162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/06/2022]
Abstract
Placental tissue intracellular calcium (Ca2+) regulates placental development and growth. Maternal high-fat diet (HFD) results in placental lipid accumulation, increased inflammation, reduced nutrient transport expression, and intrauterine growth restriction (IUGR). Currently, whether maternal HFD differentially affects placental and fetal growth and development under reduced Ca2+ influx is not yet known. We hypothesized that maternal HFD feeding decreases placental growth and development resulting in IUGR and that reduction of Ca2+ influx in the placenta worsens maternal HFD-induced placental dysfunction and IUGR. Three-week-old female B6129SF2/J wild type (WT) and transient receptor potential canonical 1 (TRPC1) protein deficient (KO) mice were fed normal fat (NF, 16 kcal % fat) and high fat (HF, 45 kcal % fat) diets for 12 weeks prior to mating with NF diet fed male mice. Fetuses and placentae were examined at mid- (D12) and late- (D18) gestation. At D12, maternal HFD had no effects on placental or fetal weight changes in WT and TRPC1 KO mice while absence of TRPC1 resulted in decreased placental and fetal weights. At D18, maternal HFD increased placental weights in both TRPC1 KO and WT mice, in part, by moderately increasing placental tissue triacylglyceride (TAG, P=.0632). At D12, mRNA expression of key placental growth factors including IGF1, PLGF, and VEGF were increased in WT compared to TRPC1 KO mice while IGF2 and VEGF mRNA expression were increased at D18. Results presented in our study demonstrated that maternal HFD increased placental weight, in part, due to increased lipid concentration resulting in IUGR and via an additive adverse effect of genotype and maternal HFD. Future studies are needed to determine the signaling mechanism underlying Ca2+ influx reduction-induced placental dysfunction and IUGR.
Collapse
|
11
|
Puspita RD, Rizal DM, Syarif RA, Sari IP. Role of COX-2 for Successful Embryo Implantation Process: A Mini-review. Open Access Maced J Med Sci 2023. [DOI: 10.3889/oamjms.2023.9123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023] Open
Abstract
The endometrium undergoes a dynamic proliferation of cells and vascular tissue under the influence of ovarian steroid hormones. Implantation is an essential process in the development of pregnancy, where there is close contact between embryo and uterus, including supposition, adhesion, and invasion. The changes occur in the human endometrium, including endometrial secretion changes, blood vessels, and immune response, leading to the uterine receptivity period. Cyclooxygenase (COX) is an enzyme that plays a role in the metabolic conversion of arachidonic acid to prostaglandins (PG). It is known that Cyclooxygenase-2 (COX-2) plays a key role in the endometrium. COX-2 is essential for blastocyst implantation and decidualization. The deficiency of COX-2, but not COX-1, results in multiple female reproductive failures (including implantation defects). We reviewed the literature on COX-2 and embryonal implantation in the endometrium and its potential mechanisms that lead to physiological implantation. This review aims to identify the essential roles of COX-2 in the successful implantation process, especially in decidualization, implantation, and embryo growth. The regulation of COX-2 expression in endometrial cells is controlled by ovarian steroid hormones (progesterone and estrogen) through the ENaC pathway to regulate the phosphorylation CREB transcription factor. The presentation of COX-2 varies throughout the stage of embryo development.
Collapse
|
12
|
Xia X, Zhang Y, Cao M, Yu X, Gao L, Qin L, Wu W, Cui Y, Liu J. Adverse effect of assisted reproductive technology-related hyperoestrogensim on the secretion and absorption of uterine fluid in superovulating mice during the peri-implantation period. Front Endocrinol (Lausanne) 2023; 14:859204. [PMID: 36950692 PMCID: PMC10027003 DOI: 10.3389/fendo.2023.859204] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/21/2023] [Indexed: 03/08/2023] Open
Abstract
OBJECTIVES This study aimed to investigate the potential mechanism of hyperoestrogensim elicited by ovulation induction affects endometrial receptivity and leads to embryo implantation abnormality or failure. STUDY DESIGN Establishment of ovulation induction mouse model. Changes in mouse body weight, ovarian weight, serum E2 level and oestrous cycle were observed. During the peri-implantation period, morphological changes in the mouse uterus and implantation sites and the localization and protein levels of oestrogen receptors ERα and ERβ, the tight junction factors CLDN3 and OCLN, the aquaporins AQP3, AQP4 and AQP8, and the sodium channel proteins SCNN1α, SCNN1β and SCNN1γ were observed. The expression and cellular localization of ERα, CLDN3, AQP8 and SCNN1 β in RL95-2 cell line were also detected by western blotting and immunofluorescence. RESULTS Ovarian and body weights were significantly higher in the 5 IU and 10 IU groups than in the CON group. The E2 level was significantly higher in the 10 IU group than in the CON group. The mice in the 10 IU group had a disordered oestrous cycle and were in oestrus for a long time. At 5.5 dpc, significantly fewer implantation sites were observed in the 10 IU group than in the CON (p<0.001) and 5 IU (p<0.05) groups. The probability of abnormal implantation and abortion was higher in the 10 IU group than in the CON and 5 IU groups. CLDN3, OCLN, AQP8 and SCNN1β in the mouse endometrium were localized on the luminal epithelium and glandular epithelium and expression levels were lower in the 10 IU group than in the CON group. The protein expression level of ERα was increased by 50% in the 10 IU group compared to the CON group. The expressions of CLDN3, AQP8, SCNN1β in RL95-2 cell line were significantly depressed by the superphysiological E2, ERα agonist or ERβ agonist, which could be reversed by the oestrogen receptor antagonist. CONCLUSION ART-induced hyperoestrogenism reduces CLDN3, AQP8 and SCNN1β expression through ERα, thereby destroying tight junctions and water and sodium channels in the endometrial cavity epithelium, which may cause abnormal implantation due to abnormal uterine fluid secretion and absorption.
Collapse
Affiliation(s)
- Xinru Xia
- State Key Laboratory of Reproductive Medicine, Center for Clinical Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Yuan Zhang
- State Key Laboratory of Reproductive Medicine, Center for Clinical Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Meng Cao
- State Key Laboratory of Reproductive Medicine, Center for Clinical Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Xiang Yu
- Department of Pediatrics, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Li Gao
- State Key Laboratory of Reproductive Medicine, Center for Clinical Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Lianju Qin
- State Key Laboratory of Reproductive Medicine, Center for Clinical Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Wei Wu
- State Key Laboratory of Reproductive Medicine, Center for Clinical Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Yugui Cui
- State Key Laboratory of Reproductive Medicine, Center for Clinical Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Jiayin Liu
- State Key Laboratory of Reproductive Medicine, Center for Clinical Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, China
- *Correspondence: Jiayin Liu,
| |
Collapse
|
13
|
Abstract
Recurrent implantation failure (RIF) is a major limiting factor in the success rates of in-vitro fertilisation despite the remarkable clinical and technological advancement made at improving assisted reproductive technology. The primary purpose of the endometrium is to provide a receptive site for the implantation of the blastocyst and support its growth and subsequent development. Endometrial pathologies such as endometrial polyps, adenomyosis, Asherman's syndrome, chronic endometritis, and congenital Müllerian ducts defect negatively influence the integrity and receptivity of the endometrium, as well as the implantation of the embryo. This review highlights the implications of these endometrial pathologies and their association with RIF.
Collapse
Affiliation(s)
| | - Babatunde Okewale
- IVF and Fertility Unit, St. Ives Specialist Hospital, Lagos, Nigeria
| |
Collapse
|
14
|
Ion Channels in Endometrial Cancer. Cancers (Basel) 2022; 14:cancers14194733. [PMID: 36230654 PMCID: PMC9564232 DOI: 10.3390/cancers14194733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Uterine or endometrial cancer is one of the most common types of cancer among the female population. Different alterations of molecules are related to many types of cancer. Some molecules called ion channels have been described as involved in the development of cancer, including endometrial cancer. We review the scientific evidence about the involvement of the ion channels in endometrial cancer and how some treatments can be developed with these molecules as a target. Even though they are involved in the progression of endometrial cancer, since they are present throughout the whole body, some possible treatments based on these could be studied. Abstract Uterine or endometrial cancer (EC) is the sixth most common neoplasia among women worldwide. Cancer can originate from a myriad of causes, and increasing evidence suggests that ion channels (IC) play an important role in the process of carcinogenesis, taking part in many pathways such as self-sufficiency in growth signals, proliferation, evasion of programmed cell death (apoptosis), angiogenesis, cell differentiation, migration, adhesion, and metastasis. Hormones and growth factors are well-known to be involved in the development and/or progression of many cancers and can also regulate some ion channels and pumps. Since the endometrium is responsive and regulated by these factors, the ICs could make an important contribution to the development and progression of endometrial cancer. In this review, we explore what is beyond (ion) flow regulation by investigating the role of the main families of ICs in EC, including as possible targets for EC treatment.
Collapse
|
15
|
Wuchu F, Ma X, Que Y, Chen J, Ruan YC. Biphasic regulation of CFTR expression by ENaC in epithelial cells: The involvement of Ca2+-modulated cAMP production. Front Cell Dev Biol 2022; 10:781762. [PMID: 36111343 PMCID: PMC9469783 DOI: 10.3389/fcell.2022.781762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 07/07/2022] [Indexed: 11/17/2022] Open
Abstract
The regulatory interaction between two typical epithelial ion channels, cystic fibrosis transmembrane conductance regulator (CFTR) and the epithelial sodium channel (ENaC), for epithelial homeostasis has been noted, although the underlying mechanisms remain unclear. Here, we report that in a human endometrial epithelial cell line (ISK), shRNA-based stable knockdown of ENaC produced a biphasic effect: a low (∼23%) degree of ENaC knockdown resulted in significant increases in CFTR mRNA and protein levels, CFTR-mediated Cl− transport activity as well as intracellular cAMP concentration, while a higher degree (∼50%) of ENaC knockdown did not further increase but restored CFTR expression and cAMP levels. The basal intracellular Ca2+ level of ISK cells was lowered by ENaC knockdown or inhibition in a degree-dependent manner. BAPTA-AM, an intracellular Ca2+ chelator that lowers free Ca2+ concentration, elevated cAMP level and CFTR mRNA expression at a low (5 µM) but not a high (50 µM) dose, mimicking the biphasic effect of ENaC knockdown. Moreover, KH-7, a selective inhibitor of soluble adenylyl cyclase (sAC), abolished the CFTR upregulation induced by low-degree ENaC knockdown or Ca2+ chelation, suggesting the involvement of sAC-driven cAMP production in the positive regulation. A luciferase reporter to indicate CFTR transcription revealed that all tested degrees of ENaC knockdown/inhibition stimulated CFTR transcription in ISK cells, suggesting that the negative regulation on CFTR expression by the high-degree ENaC deficiency might occur at post-transcription stages. Additionally, similar biphasic effect of ENaC knockdown on CFTR expression was observed in a human bronchial epithelial cell line. Taken together, these results have revealed a previously unidentified biphasic regulatory role of ENaC in tuning CFTR expression involving Ca2+-modulated cAMP production, which may provide an efficient mechanism for dynamics and plasticity of the epithelial tissues in various physiological or pathological contexts.
Collapse
Affiliation(s)
- Fulei Wuchu
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Xiyang Ma
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Yanting Que
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
| | - Junjiang Chen
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Department of Physiology, Jinan University, Guangzhou, China
| | - Ye Chun Ruan
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
- Shenzhen Research Institute, Hong Kong Polytechnic University, Shenzhen, China
- *Correspondence: Ye Chun Ruan,
| |
Collapse
|
16
|
de Souza Fonseca PA, Suárez-Vega A, Cánovas A. Unrevealing functional candidate genes for bovine fertility through RNA sequencing meta-analysis and regulatory elements networks of co-expressed genes and lncRNAs. Funct Integr Genomics 2022; 22:1361-1376. [PMID: 36001276 DOI: 10.1007/s10142-022-00893-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 07/16/2022] [Accepted: 08/09/2022] [Indexed: 11/25/2022]
Abstract
The high genetic heterogeneity and environmental effects of subfertility in livestock species make the elucidation of the genetic mechanisms associated with reproductive efficiency a difficult task. Network and co-expression network meta-analyses were applied alongside genetic variant calling and long non-coding RNA (lncRNA) characterization to identify functionally relevant target genes and regulatory subnetworks associated with fertility in dairy cattle. In total, 505 lncRNAs (441 previously annotated in the bovine reference genome ARS-UCD 1.2 and 64 novel lncRNAs) were identified. Seven differentially expressed genes between high-fertile (HF) and sub-fertile (SF) Holstein cows were identified in the network meta-analysis (CA5A, ENSBTAG00000051149, ENSBTAG00000003272, DEFB7, DIO2, TRPV3, and COL4A4). Additionally, seven functional candidate differentially co-expressed (DcoExp) modules with a differential regulatory pattern (|z-score|>2) were identified between HF and SF cows. The functional candidate genes and DcoExp modules identified were associated with fertility relevant processes such as the regulation of embryonic implantation and proliferation, interaction and molecule transfer between the fetus and the cow, and the immune system. These results help to better understand the genetic mechanisms associated with reproductive efficiency in dairy cattle through the identification of potential biomarkers and genetic variants associated with differentially expressed regulatory gene and lncRNAs regulatory element networks.
Collapse
Affiliation(s)
- Pablo Augusto de Souza Fonseca
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Aroa Suárez-Vega
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Angela Cánovas
- Centre for Genetic Improvement of Livestock, Department of Animal Biosciences, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
17
|
Reproductive Consequences of Electrolyte Disturbances in Domestic Animals. BIOLOGY 2022; 11:biology11071006. [PMID: 36101387 PMCID: PMC9312130 DOI: 10.3390/biology11071006] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/20/2022] [Accepted: 06/28/2022] [Indexed: 12/13/2022]
Abstract
Electrolyte balance is essential to maintain homeostasis in the body. The most crucial electrolytes are sodium (Na+), potassium (K+), magnesium (Mg2+), chloride (Cl−), and calcium (Ca2+). These ions maintain the volume of body fluids, and blood pressure, participate in muscle contractions, and nerve conduction, and are important in enzymatic reactions. The balance is mainly ensured by the kidneys, which are an important organ that regulates the volume and composition of urine, together with which excess electrolytes are excreted. They are also important in the reproductive system, where they play a key role. In the male reproductive system, electrolytes are important in acrosomal reaction and sperm motility. Sodium, calcium, magnesium, and chloride are related to sperm capacitation. Moreover, Mg2+, Ca2+, and Na+ play a key role in spermatogenesis and the maintenance of morphologically normal spermatozoa. Infertility problems are becoming more common. It is known that disturbances in the electrolyte balance lead to reproductive dysfunction. In men, there is a decrease in sperm motility, loss of sperm capacitation, and male infertility. In the female reproductive system, sodium is associated with estrogen synthesis. In the contraction and relaxation of the uterus, there is sodium, potassium, and calcium. Calcium is associated with oocyte activation. In turn, in women, changes in the composition of the follicular fluid are observed, leading to a restriction of follicular growth. Imbalance of oocyte electrolytes, resulting in a lack of oocyte activation and, consequently, infertility.
Collapse
|
18
|
Chen X, Fernando SR, Lee YL, Yeung WSB, Ng EHY, Li RHW, Lee KF. High-Throughput In Vitro Screening Identified Nemadipine as a Novel Suppressor of Embryo Implantation. Int J Mol Sci 2022; 23:ijms23095073. [PMID: 35563464 PMCID: PMC9103851 DOI: 10.3390/ijms23095073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/29/2022] [Accepted: 05/01/2022] [Indexed: 02/01/2023] Open
Abstract
Current contraceptive methods interfere with folliculogenesis, fertilization, and embryo implantation by physical or hormonal approaches. Although hormonal contraceptive pills are effective in regulating egg formation, they are less effective in preventing embryo implantation. To explore the use of non-hormonal compounds that suppress embryo implantation, we established a high-throughput spheroid-endometrial epithelial cell co-culture assay to screen the Library of Pharmacologically Active Compounds (LOPAC) for compounds that affect trophoblastic spheroid (blastocyst surrogate) attachment onto endometrial epithelial Ishikawa cells. We identified 174 out of 1280 LOPAC that significantly suppressed BeWo spheroid attachment onto endometrial Ishikawa cells. Among the top 20 compounds, we found the one with the lowest cytotoxicity in Ishikawa cells, P11B5, which was later identified as Nemadipine-A. Nemadipine-A at 10 µM also suppressed BeWo spheroid attachment onto endometrial epithelial RL95-2 cells and primary human endometrial epithelial cells (hEECs) isolated from LH +7/8-day endometrial biopsies. Mice at 1.5 days post coitum (dpc) treated with a transcervical injection of 100 µg/kg Nemadipine-A or 500 µg/kg PRI-724 (control, Wnt-inhibitor), but not 10 µg/kg Nemadipine-A, suppressed embryo implantation compared with controls. The transcript expressions of endometrial receptivity markers, integrin αV (ITGAV) and mucin 1 (MUC1), but not β-catenin (CTNNB1), were significantly decreased at 2.5 dpc in the uterus of treated mice compared with controls. The reduction of embryo implantation by Nemadipine-A was likely mediated through suppressing endometrial receptivity molecules ITGAV and MUC1. Nemadipine-A is a potential novel non-hormonal compound for contraception.
Collapse
Affiliation(s)
- Xian Chen
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (X.C.); (S.R.F.); (Y.-L.L.); (W.S.-B.Y.); (E.H.-Y.N.); (R.H.-W.L.)
| | - Sudini Ranshaya Fernando
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (X.C.); (S.R.F.); (Y.-L.L.); (W.S.-B.Y.); (E.H.-Y.N.); (R.H.-W.L.)
- Department of Animal Science, Faculty of Animal Science and Export Agriculture, Uva Wellassa University, Badulla 90000, Sri Lanka
| | - Yin-Lau Lee
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (X.C.); (S.R.F.); (Y.-L.L.); (W.S.-B.Y.); (E.H.-Y.N.); (R.H.-W.L.)
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518000, China
| | - William Shu-Biu Yeung
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (X.C.); (S.R.F.); (Y.-L.L.); (W.S.-B.Y.); (E.H.-Y.N.); (R.H.-W.L.)
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518000, China
| | - Ernest Hung-Yu Ng
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (X.C.); (S.R.F.); (Y.-L.L.); (W.S.-B.Y.); (E.H.-Y.N.); (R.H.-W.L.)
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518000, China
| | - Raymond Hang-Wun Li
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (X.C.); (S.R.F.); (Y.-L.L.); (W.S.-B.Y.); (E.H.-Y.N.); (R.H.-W.L.)
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518000, China
| | - Kai-Fai Lee
- Department of Obstetrics and Gynaecology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (X.C.); (S.R.F.); (Y.-L.L.); (W.S.-B.Y.); (E.H.-Y.N.); (R.H.-W.L.)
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518000, China
- Correspondence: ; Fax: +852-28161947
| |
Collapse
|
19
|
Store-Operated Ca 2+ Entry Contributes to Piezo1-Induced Ca 2+ Increase in Human Endometrial Stem Cells. Int J Mol Sci 2022; 23:ijms23073763. [PMID: 35409116 PMCID: PMC8998223 DOI: 10.3390/ijms23073763] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/21/2022] [Accepted: 03/28/2022] [Indexed: 02/04/2023] Open
Abstract
Endometrial mesenchymal stem cells (eMSCs) are a specific class of stromal cells which have the capability to migrate, develop and differentiate into different types of cells such as adipocytes, osteocytes or chondrocytes. It is this unique plasticity that makes the eMSCs significant for cellular therapy and regenerative medicine. Stem cells choose their way of development by analyzing the extracellular and intracellular signals generated by a mechanical force from the microenvironment. Mechanosensitive channels are part of the cellular toolkit that feels the mechanical environment and can transduce mechanical stimuli to intracellular signaling pathways. Here, we identify previously recorded, mechanosensitive (MS), stretch-activated channels as Piezo1 proteins in the plasma membrane of eMSCs. Piezo1 activity triggered by the channel agonist Yoda1 elicits influx of Ca2+, a known modulator of cytoskeleton reorganization and cell motility. We found that store-operated Ca2+ entry (SOCE) formed by Ca2+-selective channel ORAI1 and Ca2+ sensors STIM1/STIM2 contributes to Piezo1-induced Ca2+ influx in eMSCs. Particularly, the Yoda1-induced increase in intracellular Ca2+ ([Ca2+]i) is partially abolished by 2-APB, a well-known inhibitor of SOCE. Flow cytometry analysis and wound healing assay showed that long-term activation of Piezo1 or SOCE does not have a cytotoxic effect on eMSCs but suppresses their migratory capacity and the rate of cell proliferation. We propose that the Piezo1 and SOCE are both important determinants in [Ca2+]i regulation, which critically affects the migratory activity of eMSCs and, therefore, could influence the regenerative potential of these cells.
Collapse
|
20
|
Hou Z, He A, Zhang Q, Liu N, Liu D, Li Y, Xu B, Wang Y, Li S, Tian F, Liao T, Zhang Y, Cao J, Cao E, Li Y. Endometrial fluid aspiration immediately prior to embryo transfer does not affect IVF/vitrified-warmed embryo transfer outcomes - a prospective matched cohort study. Reprod Biomed Online 2022; 44:486-493. [PMID: 35177340 DOI: 10.1016/j.rbmo.2021.11.004] [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: 06/22/2021] [Revised: 10/21/2021] [Accepted: 11/01/2021] [Indexed: 11/29/2022]
Abstract
RESEARCH QUESTION Does the endometrial aspiration of ultrasound-invisible fluid immediately preceding embryo transfer affect IVF/vitrified-warmed embryo transfer outcomes? DESIGN A prospective matched cohort study was conducted in 96 women and 96 control participants to assess the effect on pregnancy outcomes of endometrial aspiration performed immediately before embryo transfer. This study was carried out at a university-affiliated assisted reproductive medical centre between January 2019 and December 2019. Patients were divided into two groups. The EA group had cycles with endometrial aspiration of ultrasound-invisible fluid performed before embryo transfer and the non-EA group featured cycles without endometrial aspiration. The EA group was matched by propensity score with the non-EA group in a 1:1 ratio. The EA group consisted of 99 participants before and 96 participants after propensity score matching. There were 203 and 96 participants in the non-EA group before and after propensity score matching. RESULTS No significant differences were detected in the baseline characteristics and cycle characteristics of the EA and non-EA groups. No significant between-group differences were found in reproductive outcomes in the overall population. Subgroup analysis of blastocyst transfer cycles showed the implantation rate was significantly higher in the EA group (61 women per group, 57.1% versus 40.8%, relative risk 1.40, 95% confidence interval 1.04-1.88; P = 0.022). Live birth rate, clinical pregnancy rate, ongoing pregnancy rate and multiple pregnancy rate were not different among the groups. CONCLUSIONS Endometrial aspiration immediately preceding embryo transfer does not affect IVF/vitrified-warmed embryo transfer outcomes. Interestingly, it might improve the vitrified-warmed blastocyst implantation rate. Randomized controlled trials are needed to confirm this result.
Collapse
Affiliation(s)
- Zhaojuan Hou
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, Changsha City Hunan Province, P.R. China; Clinical Research Center For Women's Reproductive Health In Hunan Province, Changsha City Hunan Province, P.R. China
| | - Aihua He
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, Changsha City Hunan Province, P.R. China; Clinical Research Center For Women's Reproductive Health In Hunan Province, Changsha City Hunan Province, P.R. China
| | - Qiong Zhang
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, Changsha City Hunan Province, P.R. China; Clinical Research Center For Women's Reproductive Health In Hunan Province, Changsha City Hunan Province, P.R. China
| | - Nenghui Liu
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, Changsha City Hunan Province, P.R. China; Clinical Research Center For Women's Reproductive Health In Hunan Province, Changsha City Hunan Province, P.R. China
| | - Donge Liu
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, Changsha City Hunan Province, P.R. China; Clinical Research Center For Women's Reproductive Health In Hunan Province, Changsha City Hunan Province, P.R. China
| | - Yumei Li
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, Changsha City Hunan Province, P.R. China; Clinical Research Center For Women's Reproductive Health In Hunan Province, Changsha City Hunan Province, P.R. China
| | - Bin Xu
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, Changsha City Hunan Province, P.R. China; Clinical Research Center For Women's Reproductive Health In Hunan Province, Changsha City Hunan Province, P.R. China
| | - Yonggang Wang
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, Changsha City Hunan Province, P.R. China; Clinical Research Center For Women's Reproductive Health In Hunan Province, Changsha City Hunan Province, P.R. China
| | - Shuyi Li
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, Changsha City Hunan Province, P.R. China; Clinical Research Center For Women's Reproductive Health In Hunan Province, Changsha City Hunan Province, P.R. China
| | - Fen Tian
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, Changsha City Hunan Province, P.R. China; Clinical Research Center For Women's Reproductive Health In Hunan Province, Changsha City Hunan Province, P.R. China
| | - Tingting Liao
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, Changsha City Hunan Province, P.R. China; Clinical Research Center For Women's Reproductive Health In Hunan Province, Changsha City Hunan Province, P.R. China
| | - Yeqing Zhang
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, Changsha City Hunan Province, P.R. China; Clinical Research Center For Women's Reproductive Health In Hunan Province, Changsha City Hunan Province, P.R. China
| | - Jianyun Cao
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, Changsha City Hunan Province, P.R. China; Clinical Research Center For Women's Reproductive Health In Hunan Province, Changsha City Hunan Province, P.R. China
| | - Exiang Cao
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, Changsha City Hunan Province, P.R. China; Clinical Research Center For Women's Reproductive Health In Hunan Province, Changsha City Hunan Province, P.R. China
| | - Yanping Li
- Department of Reproductive Medicine, Xiangya Hospital, Central South University, Changsha City Hunan Province, P.R. China; Clinical Research Center For Women's Reproductive Health In Hunan Province, Changsha City Hunan Province, P.R. China.
| |
Collapse
|
21
|
Oghbaei F, Zarezadeh R, Jafari-Gharabaghlou D, Ranjbar M, Nouri M, Fattahi A, Imakawa K. Epithelial-mesenchymal transition process during embryo implantation. Cell Tissue Res 2022; 388:1-17. [PMID: 35024964 DOI: 10.1007/s00441-021-03574-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 12/29/2021] [Indexed: 03/01/2023]
Abstract
The epithelial to mesenchymal transition (EMT) in endometrial epithelial and trophectoderm cells is essential for the progression of embryo implantation and its impairment could cause implantation failure. Therefore, EMT should be tightly regulated in both embryonic and endometrial cells during implantation. Studies reported the involvement of numerous factors in EMT regulation, including hormones, growth factors, transcription factors, microRNAs, aquaporins (AQPs), and ion channels. These factors act through different signaling pathways to affect the expression of epithelial and mesenchymal markers as well as the cellular cytoskeleton. Although the mechanisms involved in cancer cell EMT have been well studied, little is known about EMT during embryo implantation. Therefore, we comprehensively reviewed different factors that regulate the EMT, a key event required for the conceptus implantation to the endometrium.Summary sentence: Abnormal epithelial-mesenchymal transition (EMT) process within endometrial epithelial cells (EECs) or trophoblast cells can cause implantation failure. This process is regulated by various factors. Thus, the objective of this review was to summarize the effective factors on the EMT process during implantation.
Collapse
Affiliation(s)
- Farnaz Oghbaei
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Reza Zarezadeh
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Davoud Jafari-Gharabaghlou
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Minoo Ranjbar
- Department of Midwifery, Bonab Branch, Islamic Azad University, Bonab, Iran
| | - Mohammad Nouri
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Fattahi
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Kazuhiko Imakawa
- Laboratory of Molecular Reproduction, Research Institute of Agriculture, Tokai University, Kumamoto, 862-8652, Japan
| |
Collapse
|
22
|
Schaefer J, Vilos AG, Vilos GA, Bhattacharya M, Babwah AV. Uterine kisspeptin receptor critically regulates epithelial estrogen receptor α transcriptional activity at the time of embryo implantation in a mouse model. Mol Hum Reprod 2021; 27:gaab060. [PMID: 34524460 PMCID: PMC8786495 DOI: 10.1093/molehr/gaab060] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 09/07/2021] [Indexed: 12/14/2022] Open
Abstract
Embryo implantation failure is a major cause of infertility in women of reproductive age and a better understanding of uterine factors that regulate implantation is required for developing effective treatments for female infertility. This study investigated the role of the uterine kisspeptin receptor (KISS1R) in the molecular regulation of implantation in a mouse model. To conduct this study, a conditional uterine knockout (KO) of Kiss1r was created using the Pgr-Cre (progesterone receptor-CRE recombinase) driver. Reproductive profiling revealed that while KO females exhibited normal ovarian function and mated successfully to stud males, they exhibited significantly fewer implantation sites, reduced litter size and increased neonatal mortality demonstrating that uterine KISS1R is required for embryo implantation and a healthy pregnancy. Strikingly, in the uterus of Kiss1r KO mice on day 4 (D4) of pregnancy, the day of embryo implantation, KO females exhibited aberrantly elevated epithelial ERα (estrogen receptor α) transcriptional activity. This led to the temporal misexpression of several epithelial genes [Cftr (Cystic fibrosis transmembrane conductance regulator), Aqp5 (aquaporin 5), Aqp8 (aquaporin 8) and Cldn7 (claudin 7)] that mediate luminal fluid secretion and luminal opening. As a result, on D4 of pregnancy, the lumen remained open disrupting the final acquisition of endometrial receptivity and likely accounting for the reduction in implantation events. Our data clearly show that uterine KISS1R negatively regulates ERα signaling at the time of implantation, in part by inhibiting ERα overexpression and preventing detrimentally high ERα activity. To date, there are no reports on the regulation of ERα by KISS1R; therefore, this study has uncovered an important and powerful regulator of uterine ERα during early pregnancy.
Collapse
Affiliation(s)
- Jennifer Schaefer
- Laboratory of Human Growth and Reproductive Development, Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
- School of Graduate Studies, Joint Graduate Program in Toxicology, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Angelos G Vilos
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynaecology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - George A Vilos
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynaecology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Moshmi Bhattacharya
- Department of Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
- Child Health Institute of New Jersey, New Brunswick, NJ, USA
| | - Andy V Babwah
- Laboratory of Human Growth and Reproductive Development, Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ, USA
- School of Graduate Studies, Joint Graduate Program in Toxicology, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
- Child Health Institute of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|
23
|
Expression of SGLT1 in the Mouse Endometrial Epithelium and its Role in Early Embryonic Development and Implantation. Reprod Sci 2021; 28:3094-3108. [PMID: 34460091 DOI: 10.1007/s43032-021-00480-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/31/2021] [Indexed: 10/20/2022]
Abstract
Many functional activities of endometrium epithelium are energy consuming which are very important for maintaining intrauterine environment needed by early embryonic development and establishment of implantation window. Glucose is a main energy supplier and one of the main components of intrauterine fluid. Obviously, glucose transports in endometrium epithelium involve in for these activities but their functions have not been elucidated. In this research, we observed a spatiotemporal pattern of sodium glucose transporter 1 (SGLT1) expression in the mouse endometrium. We also determined that progesterone can promote the expression of SGLT1 in the mouse endometrial epithelium in response to the action of oestrogen. Treatment with the SGLT1 inhibitor phlorizin or small interfering RNA specific for SGLT1 (SGLT1-siRNA) altered glucose uptake in primary cultured endometrial epithelial cells, which exhibited reduced ATP levels and AMPK activation. The injection of phlorizin or SGLT1-siRNA into one uterine horn of each mouse on day 2 of pregnancy led to an increased glucose concentration in the uterine fluid and decreased number of harvested normal blastocysts and decreased expression of integrin αVβ3 in endometrial epithelium and increased expression of mucin 1 and lactoferrin in endometrial epithelium and the uterine homogenates exhibited activated AMPK, a decreased ATP level on day 4, and a decreased number of implantation sites on day 5. In embryo transfer experiments, pre-treatment of the uterine horn with phlorizin or SGLT1-siRNA during the implantation window led to a decreased embryo implantation rate on day 5 of pregnancy, even when embryos from normal donor mice were used. In conclusion, SGLT1, which participates in glucose transport in the mouse endometrial epithelium, inhibition and/or reduced expression of SGLT1 affects early embryo development by altering the glucose concentration in the uterine fluid. Inhibition and/or reduced expression of SGLT1 also affects embryo implantation by influencing energy metabolism in epithelial cells, which consequently influences implantation-related functional activities.
Collapse
|
24
|
Zhang Y, Du X, Chen X, Tang H, Zhou Q, He J, Ding Y, Wang Y, Liu X, Geng Y. Rictor/mTORC2 is involved in endometrial receptivity by regulating epithelial remodeling. FASEB J 2021; 35:e21731. [PMID: 34131963 DOI: 10.1096/fj.202100529rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 11/11/2022]
Abstract
Successful embryo implantation requires well-functioning endometrial luminal epithelial cells to establish uterine receptivity. Inadequate uterine receptivity is responsible for approximately two thirds of implantation failures in humans. However, the regulatory mechanism governing this functional process remains largely unexplored. A previous study revealed that the expression of Rictor, the main member of mTORC2, in mouse epithelial cells is increased on the fourth day of gestation (D4). Here, we provide the first report of the involvement of Rictor in the regulation of endometrial receptivity. Rictor was conditionally ablated in the mouse endometrium using a progesterone receptor cre (PRcre ) mouse model. Loss of Rictor altered polarity remodeling and the Na+ channel protein of endometrial cells by mediating Rac-1/PAK1(pPAK1)/ERM(pERM) and Sgk1/pSgk1 signaling, respectively, ultimately resulting in impaired fertility. In the endometrium of women with infertility, the expression of Rictor was changed, along with the morphological transformation and Na+ channel protein of epithelial cells. Our findings demonstrate that Rictor is crucial for the establishment of uterine receptivity in both mice and humans. The present study may help improve the molecular regulatory network of endometrial receptivity and provide new diagnostic and treatment strategies for infertility.
Collapse
Affiliation(s)
- Yue Zhang
- Joint International Research Laboratory of Reproduction & Development, School of Public Health and Management, Chongqing Medical University, Chongqing, P.R. China
| | - Xinman Du
- Joint International Research Laboratory of Reproduction & Development, School of Public Health and Management, Chongqing Medical University, Chongqing, P.R. China
| | - Xuemei Chen
- Joint International Research Laboratory of Reproduction & Development, School of Public Health and Management, Chongqing Medical University, Chongqing, P.R. China
| | - Hongyu Tang
- Joint International Research Laboratory of Reproduction & Development, School of Public Health and Management, Chongqing Medical University, Chongqing, P.R. China
| | - Qin Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Junlin He
- Joint International Research Laboratory of Reproduction & Development, School of Public Health and Management, Chongqing Medical University, Chongqing, P.R. China
| | - Yubin Ding
- Joint International Research Laboratory of Reproduction & Development, School of Public Health and Management, Chongqing Medical University, Chongqing, P.R. China
| | - Yingxiong Wang
- Joint International Research Laboratory of Reproduction & Development, School of Public Health and Management, Chongqing Medical University, Chongqing, P.R. China
- College of Basic Medicine, Chongqing Medical University, Chongqing, P.R. China
| | - Xueqing Liu
- Joint International Research Laboratory of Reproduction & Development, School of Public Health and Management, Chongqing Medical University, Chongqing, P.R. China
| | - Yanqing Geng
- Joint International Research Laboratory of Reproduction & Development, School of Public Health and Management, Chongqing Medical University, Chongqing, P.R. China
- College of Basic Medicine, Chongqing Medical University, Chongqing, P.R. China
| |
Collapse
|
25
|
Wang D, Li W, Yang C, Chen X, Liu X, He J, Tong C, Peng C, Ding Y, Geng Y, Cao X, Li F, Gao R, Wang Y. Exposure to ethylparaben and propylparaben interfere with embryo implantation by compromising endometrial decidualization in early pregnant mice. J Appl Toxicol 2021; 41:1732-1746. [PMID: 34101200 DOI: 10.1002/jat.4208] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 05/03/2021] [Accepted: 05/24/2021] [Indexed: 11/10/2022]
Abstract
Ethylparaben (EtP) and propylparaben (PrP) are common preservatives and well-known endocrine-disrupting chemicals. Studies have demonstrated that they can reduce female fertility, but the underlying mechanism, especially that on embryo implantation, is still poorly understood. Endometrial decidualization is a critical event for embryo implantation. In this study, we aimed to explore the effects of EtP/PrP on endometrial decidualization. Pregnant mice were dosed daily by oral gavage with EtP at 0, 400, 800 and 1600 mg/kg or with PrP at 0, 625, 1250 and 2500 mg/kg from Day 1 of pregnancy until sacrifice. The results showed that the rate of pregnant mice with impaired embryo implantation, whose number of implantation sites was less than 7, was significantly increased after exposure to 1600 mg/kg EtP or 2500 mg/kg PrP. Further study found that the expression of endometrial decidualization markers HOXA10, MMP9 and PR was significantly downregulated in 1600 mg/kg EtP group and 2500 mg/kg PrP group. Notably, serum oestrogen and progesterone levels were significantly increased, whereas the expression of uterine oestrogen receptor and progesterone receptor was decreased following 1600 mg/kg EtP or 2500 mg/kg PrP exposure. In the breeding test, fewer offspring were found after females were exposed to 1600 mg/kg EtP or 2500 mg/kg PrP in early pregnancy. This demonstrated that exposure to EtP/PrP interfered with embryo implantation by compromising endometrial decidualization in early-stage pregnant mice. Disorders of reproductive hormones and hormone receptor signals could be responsible for impaired decidualization. This study broadened the understanding on the biological safety of EtP and PrP.
Collapse
Affiliation(s)
- Dan Wang
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Weike Li
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Chengshun Yang
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Xuemei Chen
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Xueqing Liu
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Junlin He
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Chao Tong
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China.,Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chuan Peng
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China.,The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yubin Ding
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Yanqing Geng
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Xianqing Cao
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Fangfang Li
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Rufei Gao
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| | - Yingxiong Wang
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, China
| |
Collapse
|
26
|
Li L, Zhang W, Tong C(L, Yan H, Yin P, Li K, Heng BC, Tong G. Differential cytokines expression in cervical exfoliated cells of women with implantation after in vitro fertilization. Acta Biochim Biophys Sin (Shanghai) 2020; 52:1281-1284. [PMID: 33175952 DOI: 10.1093/abbs/gmaa103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 07/02/2020] [Indexed: 11/14/2022] Open
Affiliation(s)
- Li Li
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | | | - Christina (Ling) Tong
- School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA, USA
| | - Hua Yan
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ping Yin
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Kai Li
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Boon Chin Heng
- School of Stomatology, Peking University, Beijing 100081, China
| | - Guoqing Tong
- Reproductive Medicine Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
27
|
Hernández-Vargas P, Muñoz M, Domínguez F. Identifying biomarkers for predicting successful embryo implantation: applying single to multi-OMICs to improve reproductive outcomes. Hum Reprod Update 2020; 26:264-301. [PMID: 32096829 DOI: 10.1093/humupd/dmz042] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 10/08/2019] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Successful embryo implantation is a complex process that requires the coordination of a series of events, involving both the embryo and the maternal endometrium. Key to this process is the intricate cascade of molecular mechanisms regulated by endocrine, paracrine and autocrine modulators of embryonic and maternal origin. Despite significant progress in ART, implantation failure still affects numerous infertile couples worldwide and fewer than 10% of embryos successfully implant. Improved selection of both the viable embryos and the optimal endometrial phenotype for transfer remains crucial to enhancing implantation chances. However, both classical morphological embryo selection and new strategies incorporated into clinical practice, such as embryonic genetic analysis, morphokinetics or ultrasound endometrial dating, remain insufficient to predict successful implantation. Additionally, no techniques are widely applied to analyse molecular signals involved in the embryo-uterine interaction. More reliable biological markers to predict embryo and uterine reproductive competence are needed to improve pregnancy outcomes. Recent years have seen a trend towards 'omics' methods, which enable the assessment of complete endometrial and embryonic molecular profiles during implantation. Omics have advanced our knowledge of the implantation process, identifying potential but rarely implemented biomarkers of successful implantation. OBJECTIVE AND RATIONALE Differences between the findings of published omics studies, and perhaps because embryonic and endometrial molecular signatures were often not investigated jointly, have prevented firm conclusions being reached. A timely review summarizing omics studies on the molecular determinants of human implantation in both the embryo and the endometrium will help facilitate integrative and reliable omics approaches to enhance ART outcomes. SEARCH METHODS In order to provide a comprehensive review of the literature published up to September 2019, Medline databases were searched using keywords pertaining to omics, including 'transcriptome', 'proteome', 'secretome', 'metabolome' and 'expression profiles', combined with terms related to implantation, such as 'endometrial receptivity', 'embryo viability' and 'embryo implantation'. No language restrictions were imposed. References from articles were also used for additional literature. OUTCOMES Here we provide a complete summary of the major achievements in human implantation research supplied by omics approaches, highlighting their potential to improve reproductive outcomes while fully elucidating the implantation mechanism. The review highlights the existence of discrepancies among the postulated biomarkers from studies on embryo viability or endometrial receptivity, even using the same omic analysis. WIDER IMPLICATIONS Despite the huge amount of biomarker information provided by omics, we still do not have enough evidence to link data from all omics with an implantation outcome. However, in the foreseeable future, application of minimally or non-invasive omics tools, together with a more integrative interpretation of uniformly collected data, will help to overcome the difficulties for clinical implementation of omics tools. Omics assays of the embryo and endometrium are being proposed or already being used as diagnostic tools for personalised single-embryo transfer in the most favourable endometrial environment, avoiding the risk of multiple pregnancies and ensuring better pregnancy rates.
Collapse
Affiliation(s)
- Purificación Hernández-Vargas
- IVI-RMA Alicante, Innovation. Avda. de Denia 111, 03015 Alicante, Spain.,Fundación IVI, Innovation-IIS La Fe, Avda. Fernando Abril Martorell 106, Torre A, 1° 1.23, 46026 Valencia, Spain
| | - Manuel Muñoz
- IVI-RMA Alicante, Innovation. Avda. de Denia 111, 03015 Alicante, Spain.,Fundación IVI, Innovation-IIS La Fe, Avda. Fernando Abril Martorell 106, Torre A, 1° 1.23, 46026 Valencia, Spain
| | - Francisco Domínguez
- Fundación IVI, Innovation-IIS La Fe, Avda. Fernando Abril Martorell 106, Torre A, 1° 1.23, 46026 Valencia, Spain
| |
Collapse
|
28
|
Zarei R, Nikpour P, Rashidi B, Eskandari N, Aboutorabi R. Evaluation of Muc1 Gene Expression at The Time of Implantation in Diabetic Rat Models Treated with Insulin, Metformin and Pioglitazone in The Normal Cycle and Ovulation Induction Cycle. INTERNATIONAL JOURNAL OF FERTILITY & STERILITY 2020; 14:218-222. [PMID: 33098389 PMCID: PMC7604705 DOI: 10.22074/ijfs.2020.44409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 08/02/2020] [Indexed: 12/25/2022]
Abstract
Background Mucin-1(Muc1) is one of the first molecules in the endometrium that confronts implanting embryos. There is insufficient knowledge about the impacts of diabetes and drugs developed for diabetes treatment on expression of this molecule at the time of implantation. Therefore, this study aimed to investigate the impacts of diabetes and insulin, metformin and pioglitazone on Muc1 expression at the time of implantation. Materials and Methods This experimental study was conducted on a total of 63 female Wistar rats divided into 9 groups. To induce type 1diabetes, streptozotocin (STZ) and for induction of type 2 diabetes, nicotinamide (NA) and STZ were injected intraperitoneally. For superovulation, human menopausal gonadotropin (HMG) and human chorionic gonadotropin (HCG) were used. Insulin, metformin and pioglitazone were administered for two weeks. Finally, the endometrial expression of Muc1 was evaluated by quantitative real-time reverse transcription-polymerase chain reaction (RT-PCR). Results Muc1 expression was non-significantly increased in type 1 and type 2 diabetic groups compared to the control group (P=0.61 and 0.13, respectively); also, it increased in insulin-treated type 1 diabetic group compared to the control group (P=0.0001). Its expression was increased in insulin-treated type 1 diabetic group compared to untreated diabetic group (P=0.001). The expression level of Muc1 was significantly reduced in superovulated and insulintreated type 1 diabetic group compared to the insulin-treated type 1 diabetic group (P=0.001). Conclusion One of the causes of fertility problems in diabetes, is changes in Muc1 expression during implantation. On the other hand, the use of insulin in these patients can even lead to overexpression of this gene and worsen the condition. However, these changes can be partially mitigated by assisted reproductive technology (ART) such as superovulation. Also, treatment with metformin and pioglitazone can restore Muc1 expression to near normal levels and has beneficial effects on implantation.
Collapse
Affiliation(s)
- Ronak Zarei
- Department of Anatomical Sciences, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parvaneh Nikpour
- Department of Genetics and Molecular Biology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.,Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Bahman Rashidi
- Department of Anatomical Sciences, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nahid Eskandari
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Roshanak Aboutorabi
- Department of Anatomical Sciences, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran. Electronic Address:
| |
Collapse
|
29
|
Zhang Q, Ni T, Dang Y, Ding L, Jiang J, Li J, Xia M, Yu N, Ma J, Yan J, Chen ZJ. MiR-148a-3p may contribute to flawed decidualization in recurrent implantation failure by modulating HOXC8. J Assist Reprod Genet 2020; 37:2535-2544. [PMID: 32772270 DOI: 10.1007/s10815-020-01900-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 07/21/2020] [Indexed: 10/23/2022] Open
Abstract
PURPOSE To evaluate whether miR-148a-3p overexpression is associated with disrupted decidualization of recurrent implantation failure (RIF). METHODS Endometrial miRNA and mRNA expression profiles during the implantation window derived from women with and without RIF were identified using microarray and RT-qPCR. Immortalized human endometrial stromal cells (HESCs) were cultured for proliferation and in vitro decidualization assays after enhancing miR-148a-3p expression or inhibiting putative target gene homeobox C8 (HOXC8) expression. RT-qPCR, western blot, and luciferase reporter assays were used to confirm the relationship between miR-148a-3p and HOXC8 gene. RESULTS MiR-148a-3p was significantly upregulated in RIF endometrial tissues. Forced expression of miR-148a-3p notably attenuated HESC in vitro decidualization. Mechanistic studies revealed that miR-148a-3p directly bounds to the HOXC8 3' untranslated region (3'UTR) and suppressed HOXC8 expressions in both mRNA and protein levels. Further investigations demonstrated that inhibition of HOXC8 in HESCs induced similar effects on decidual process as those induced by miR-148a-3p overexpression. CONCLUSION Taken together, our findings suggested that elevated miR-148a-3p might account for flawed decidualization in RIF by negatively regulating HOXC8, raising the possibility that miR-148a-3p might be a novel therapeutic target in RIF.
Collapse
Affiliation(s)
- Qian Zhang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, No. 157 Jingliu Road, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.,Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
| | - Tianxiang Ni
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, No. 157 Jingliu Road, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.,Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
| | - Yujie Dang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, No. 157 Jingliu Road, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.,Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
| | - Lingling Ding
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, No. 157 Jingliu Road, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.,Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
| | - Jingjing Jiang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, No. 157 Jingliu Road, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.,Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
| | - Jing Li
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, No. 157 Jingliu Road, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.,Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
| | - Mingdi Xia
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, No. 157 Jingliu Road, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.,Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
| | - Na Yu
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, No. 157 Jingliu Road, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.,Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
| | - Jinlong Ma
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, No. 157 Jingliu Road, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.,Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
| | - Junhao Yan
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, No. 157 Jingliu Road, Jinan, 250012, Shandong, China. .,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China. .,Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China. .,Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, 250012, Shandong, China.
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, No. 157 Jingliu Road, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.,Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Medicine Research Center for Reproductive Health, Shandong University, Jinan, 250012, Shandong, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetic, Shanghai, 200127, China.,Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| |
Collapse
|
30
|
Nelson W, Adu-Gyamfi EA, Czika A, Wang YX, Ding YB. Bisphenol A-induced mechanistic impairment of decidualization. Mol Reprod Dev 2020; 87:837-842. [PMID: 32691498 DOI: 10.1002/mrd.23400] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 07/06/2020] [Indexed: 12/11/2022]
Abstract
Decidualization is a crucial precedent to embryo implantation, as its impairment is a major contributor to female infertility and pregnancy complications. Unraveling the molecular mechanisms involved in the impairment of decidualization has been a subject of interest in the field of reproductive medicine. Evidence from several experimental settings show that exposure to bisphenol A (BPA), an endocrine-disrupting chemical, affects the expression of several molecules that are involved in decidualization. Both low and high doses of BPA impair decidualization through the dysregulation of estrogen (ER) and progesterone (PR) receptors. Exposure to low doses of BPA leads to decreased levels and activities of several antioxidant enzymes, increased activity of endothelial nitric oxide synthase (eNOS), and increased production of nitric oxide (NO) via the upregulation of ER and PR. Consequently, oxidative stress is induced and decidualization becomes impaired. On the other hand, exposure to high doses of BPA downregulates ER and PR and impairs decidualization through two distinct pathways. One is through the upregulation of early growth response-1 (EGR1) via increased phosphorylation of extracellular signal-regulated protein kinases 1 and 2; and the other is through a reduced serum glucocorticoid-induced kinase-1 (SGK1)-mediated downregulation of epithelial sodium channel-α and the induction of oxidative stress. Thus, regardless of the dose, BPA can impair decidualization to trigger infertility and pregnancy complications. This warrants the need to adopt lifestyles that will decrease the tendency of getting exposed to BPA.
Collapse
Affiliation(s)
- William Nelson
- Joint International Research Laboratory of Reproduction and Development, Department of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China.,Department of Environmental and Occupational Health, Muhimbili University of Health and Allied Sciences, Dar es salaam, Tanzania
| | - Enoch Appiah Adu-Gyamfi
- Joint International Research Laboratory of Reproduction and Development, Department of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Armin Czika
- Joint International Research Laboratory of Reproduction and Development, Department of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Ying-Xiong Wang
- Joint International Research Laboratory of Reproduction and Development, Department of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Yu-Bin Ding
- Joint International Research Laboratory of Reproduction and Development, Department of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, China
| |
Collapse
|
31
|
Cao J, Liu D, Zhao S, Yuan L, Huang Y, Ma J, Yang Z, Shi B, Wang L, Wei J. Estrogen attenuates TGF-β1-induced EMT in intrauterine adhesion by activating Wnt/β-catenin signaling pathway. ACTA ACUST UNITED AC 2020; 53:e9794. [PMID: 32638833 PMCID: PMC7346761 DOI: 10.1590/1414-431x20209794] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 05/22/2020] [Indexed: 12/19/2022]
Abstract
Although estrogen has crucial functions for endometrium growth, the specific dose
and underlying molecular mechanism in intrauterine adhesion (IUA) remain
unclear. In this study, we aimed to investigate the effects of estrogen on
epithelial-mesenchymal transition (EMT) in normal and fibrotic endometrium, and
the role of estrogen and Wnt/β-catenin signaling in the formation of endometrial
fibrosis. CCK-8 and immunofluorescence assay were performed to access the
proliferation of different concentrations of estrogen on normal human
endometrial epithelial cells (hEECs). qRT-PCR and western blot assay were
utilized to explore the effect of estrogen on EMT in normal and fibrotic
endometrium, and main components of Wnt/β-catenin signaling pathway in
vitro. Hematoxylin and eosin and Masson staining were used to
evaluate the effect of estrogen on endometrial morphology and fibrosis
in vivo. Our results indicated that the proliferation of
normal hEECs was inhibited by estrogen at a concentration of 30 nM accompanied
by upregulation of mesenchymal markers and downregulation of epithelial markers.
Interestingly, in the model of transforming growth factor β1 (TGF-β1)-induced
endometrial fibrosis, the same concentration of estrogen inhibited the process
of EMT, which might be partially mediated by regulation of the Wnt/β-catenin
pathway. In addition, relatively high doses of estrogen efficiently increased
the number of endometrial glands and reduced the area of fibrosis as determined
by the reduction of EMT in IUA animal models. Taken together, our results
demonstrated that an appropriate concentration of estrogen may prevent the
occurrence and development of IUA by inhibiting the TGF-β1-induced EMT and
activating the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Jia Cao
- College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Dan Liu
- College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Shiyun Zhao
- College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Liwei Yuan
- College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yani Huang
- College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Jingwen Ma
- Department of Gynecology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Zhijuan Yang
- Department of Gynecology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Bin Shi
- Department of Beijing National Biochip Research Center Sub-Center in Ningxia, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Libin Wang
- College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| | - Jun Wei
- College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
32
|
Mokhtar MH, Giribabu N, Salleh N. Testosterone Reduces Tight Junction Complexity and Down-regulates Expression of Claudin-4 and Occludin in the Endometrium in Ovariectomized, Sex-steroid Replacement Rats. In Vivo 2020; 34:225-231. [PMID: 31882482 DOI: 10.21873/invivo.11764] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 11/07/2019] [Accepted: 11/12/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND/AIM It was hypothesized that endometrial tight junction morphology and expression of tight junction proteins i.e., claudin-4 and occludin in the uterus, are affected by testosterone. Therefore, the effects of testosterone on these parameters in the uterus during receptivity period were investigated. MATERIALS AND METHODS Ovariectomized adult female rats were given testosterone (1 mg/kg/day) alone or in combination with flutamide or finasteride between days 6 to 8 of sex-steroid replacement treatment, which was considered the period of uterine receptivity. Ultramorphology of tight junctions was visualized by transmission electron microscopy while distribution and expression of claudin-4 and occludin were examined by immunofluorescence and real-time polymerase chain reaction respectively. RESULTS Administration of testosterone caused loss of tight junction complexity and down-regulated expression of claudin-4 and occludin in the uterus. CONCLUSION Decreased endometrial tight junction complexity and expression of claudin-4 and occludin in the uterus during receptivity period by testosterone may interfere with embryo attachment and subsequent implantation.
Collapse
Affiliation(s)
- Mohd Helmy Mokhtar
- Department of Physiology, Faculty of Medicine, University Kebangsaan Malaysia (UKM), Kuala Lumpur, Malaysia
| | - Nelli Giribabu
- Department of Physiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Naguib Salleh
- Department of Physiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
33
|
Identification of Differentially Expressed Gene Transcripts in Porcine Endometrium during Early Stages of Pregnancy. Life (Basel) 2020; 10:life10050068. [PMID: 32429378 PMCID: PMC7281126 DOI: 10.3390/life10050068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/13/2020] [Accepted: 05/13/2020] [Indexed: 12/13/2022] Open
Abstract
During the early stages of pregnancy, the uterine endometrium undergoes dramatic morphologic and functional changes accompanied with dynamic variation in gene expression. Pregnancy-stage specific differentially expressed gene (DEG)-transcript-probes were investigated and identified by comparing endometrium transcriptome at 9th day (9D), 12th day (12D) and 16th day (16D) of early pregnancy in Polish large-white (PLW) gilts. Endometrium comparisons between 9D-vs-12D, 9D-vs-16D and 12D-vs-16D of early pregnancy identified 6049, 374 and 6034 highly significant DEG-transcript-probes (p < 0.001; >2 FC). GO term enrichment analysis identified commonly shared upregulated endometrial DEG-transcript-probes (p < 0.001; >2 FC), that were regulating the gene functions of anatomic structure development and transport (TG), DNA-binding and methyltransferase activity (ZBTB2), ion-binding and kinase activity (CKM), cell proliferation and apoptosis activity (IL1B). Downregulated DEG-transcript-probes (p < 0.001; >2 FC) were involved in regulating the gene functions of phosphatase activity (PTPN11), TC616413 gene-transcript and Sus-scrofa LOC100525539. Moreover, blastn comparison of microarray-probes sequences against sus-scrofa11 assembly identified commonly shared upregulated endometrial DEG-transcript-probes (E < 0.06; >2 FC), that were regulating the gene functions of reproduction and growth (SELENOP), cytoskeleton organization and kinase activity (CDC42BPA), phosphatase activity (MINPP1), enzyme-binding and cell-population proliferation (VAV3), cancer-susceptibility candidate gene (CASC4), cytoskeletal protein-binding (COBLL1), ion-binding, enzyme regulator activity (ACAP2) Downregulated endometrial DEG-transcript-probes (E < 0.06; >2FC) were involved in regulating the gene functions of signal-transduction (TMEM33), catabolic and metabolic processes (KLHL15). Microarray validation experiment on selected candidate genes showed complementarity to significant endometrial DEG-transcript-probes responsible for the regulation of immune response (IL1B, S100A11), lipid metabolism (FABP3, PPARG), cell-adhesion (ITGAV), angiogenesis (IL1B), intercellular transmission (NMB), cell-adhesion (OPN) and response to stimuli (RBP4) was confirmed by RT-PCR. This study provides a clue that identified pregnancy-stage specific microarray transcript probes could be considered as candidate genes for recognition and establishment of early pregnancy in the pig.
Collapse
|
34
|
Su Y, Zhang JJ, He JL, Liu XQ, Chen XM, Ding YB, Tong C, Peng C, Geng YQ, Wang YX, Gao RF. Endometrial autophagy is essential for embryo implantation during early pregnancy. J Mol Med (Berl) 2020; 98:555-567. [PMID: 32072231 DOI: 10.1007/s00109-019-01849-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 10/16/2019] [Accepted: 10/25/2019] [Indexed: 12/11/2022]
Abstract
Embryo implantation is an essential and complex process in mammalian reproduction. However, little evidence has indicated the involvement of autophagy during embryo implantation. To determine the possible role of autophagy in uterine of pregnant mice during the peri-implantation stage, we first examined the expression of autophagy-related markers ATG5 and LC3 on day 4, 5, and 6 of pregnancy (D4, D5, and D6, respectively). Compared with expression on D4, downregulation of the autophagy-related markers was observed on D5 and D6, the days after the embryo attached to the receptivity endometrium. Further examination showed that autophagy-related markers ATG5, ATG12, LC3, cathepsin B, and P62 at the implantation site were significantly decreased when comparing with the inter-implantation site. Fewer number of autophagosomes at the implantation site were also observed by transmission electron microscopy. To confirm the functional role of autophagy during embryo implantation in mice, we administered the autophagy inhibitor 3-methyladenine and chloroquine to mice. After treated with 3-methyladenine, the expression of decidual markers HOXA10 and progesterone receptor were significantly reduced. Furthermore, a reduction in implantation sites and increase in the HOXA10 and PR protein levels were observed in response to chloroquine treatment. In addition, impaired uterine decidualization and dysregulation of the PR and HOXA10 protein levels was observed after autophagy inhibited by 3-methyladenine and chloroquine in in vivo artificial decidualization mouse model. In the last, LC3 and P62 were also observed in normal human proliferative, secretory, and decidua tissues. In conclusion, endometrial autophagy may be essential for embryo implantation, and it may be associated with endometrial decidualization during early pregnancy. KEY MESSAGE: • Autophagy-related markers were significantly decreased at implantation site. • Autophagy inhibition results in abnormal decidualization. • Autophagy is essential for embryo implantation.
Collapse
Affiliation(s)
- Yan Su
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, 400016, China
- Joint International Research Laboratory of Reproduction&Development, Chongqing, 400016, China
| | - Juan-Juan Zhang
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, 400016, China
- Reproductive Medicine Centre, Taihe Hospital, Hubei University of Medicine, 32 South Renmin Road, Shiyan, 442000, Hubei, China
| | - Jun-Lin He
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, 400016, China
- Joint International Research Laboratory of Reproduction&Development, Chongqing, 400016, China
| | - Xue-Qing Liu
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, 400016, China
- Joint International Research Laboratory of Reproduction&Development, Chongqing, 400016, China
| | - Xue-Mei Chen
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, 400016, China
- Joint International Research Laboratory of Reproduction&Development, Chongqing, 400016, China
| | - Yu-Bin Ding
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, 400016, China
- Joint International Research Laboratory of Reproduction&Development, Chongqing, 400016, China
| | - Chao Tong
- Joint International Research Laboratory of Reproduction&Development, Chongqing, 400016, China
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Chuan Peng
- Joint International Research Laboratory of Reproduction&Development, Chongqing, 400016, China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yan-Qing Geng
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, 400016, China
- Joint International Research Laboratory of Reproduction&Development, Chongqing, 400016, China
| | - Ying-Xiong Wang
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, 400016, China.
- Joint International Research Laboratory of Reproduction&Development, Chongqing, 400016, China.
| | - Ru-Fei Gao
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing, 400016, China.
- Joint International Research Laboratory of Reproduction&Development, Chongqing, 400016, China.
| |
Collapse
|
35
|
Panigrahi M, Kumar H, Sah V, Dillipkumar Verma A, Bhushan B, Parida S. Transcriptome profiling of buffalo endometrium reveals molecular signature distinct to early pregnancy. Gene 2020; 743:144614. [PMID: 32222532 DOI: 10.1016/j.gene.2020.144614] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 03/22/2020] [Accepted: 03/23/2020] [Indexed: 12/18/2022]
Abstract
Buffalo reproduction struggles with a high incidence of early embryonic mortality. Effective treatment and prevention strategies for this condition are not available due to lack of understanding of molecular pathways in early pregnancy of this species. In the present study, we have attempted to understand these molecular pathways by characterizing the endometrial transcriptomic profiles of pregnant buffalos during early pregnancy. For the transcriptome profiling, buffalo endometrial tissues of 29-36 days of pregnancy and of nonpregnant luteal phase were collected from the local slaughterhouse. We confirmed the status of pregnancy based on the crown vertebral length of the foetus. Total RNA was isolated and sequencing was performed using the Illumina nextseq platform. The raw reads were filtered and mapped to the Bos taurus UMD 3.1 reference genome assembly. An average of 24,597 genes was investigated for differential expression between the two groups. Transcriptome data identified a total of 450 differentially expressed genes (using a cut off value of log2 fold changes >2 and <-2) in early pregnancy in comparison to the nonpregnant group (Padj < 0.05). Among these, 270 genes were significantly upregulated and 180 genes were downregulated. The most impacted pathways were related to secretion, transport, ionic homeostasis, mitosis and negative regulation of viral processes. In conclusion, our study characterized a unique set of DEGs, during the early pregnancy of buffalo, which potentially modulate the endometrial environment to establish and maintain a successful pregnancy.
Collapse
Affiliation(s)
- Manjit Panigrahi
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, UP, India
| | - Harshit Kumar
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, UP, India
| | - Vaishali Sah
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, UP, India
| | - Ankita Dillipkumar Verma
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, UP, India
| | - Bharat Bhushan
- Division of Animal Genetics, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, UP, India
| | - Subhashree Parida
- Division of Pharmacology & Toxicology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, UP, India.
| |
Collapse
|
36
|
Ran J, Yang HH, Huang HP, Huang HL, Xu Z, Zhang W, Wang ZX. ZEB1 modulates endometrial receptivity through epithelial-mesenchymal transition in endometrial epithelial cells in vitro. Biochem Biophys Res Commun 2020; 525:699-705. [PMID: 32139122 DOI: 10.1016/j.bbrc.2020.02.153] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 02/25/2020] [Indexed: 01/18/2023]
Abstract
Zinc finger E-box binding homeobox 1 (ZEB1) promotes epithelial-mesenchymal transition (EMT) in carcinogenesis, but its role in embryo implantation has not yet been identified. The present study sought to verify if ZEB1 plays a role in endometrial receptivity through regulation of EMT during embryo implantation. Endometrial epithelium from sixty patients in phase of the menstrual cycle (including proliferative and secretory phases) were collected for assessment of mRNA/protein expression. In human endometrial adenocarcinoma cell line RL95-2, ZEB1 expression was suppressed by using shRNA, and the cell function and mRNA/protein expression were evaluated. RL95-2 cells and human choriocarcinoma cell line JAR were co-cultured to establish embryo implantation model in vitro. The results showed that, ZEB1 was highly expressed at both mRNA and protein levels in human endometrium during mid-secretory phase of the menstrual cycle. Knockdown of ZEB1 expression in RL95-2 cells attenuated cell growth, migration, DNA replication, and altered expression of E-cadherin and vimentin at both mRNA and protein levels. Interestingly, knockdown of ZEB1 expression in RL95-2 cells potently suppressed JAR spheroid attachment in vitro (P < 0.01). Additionally, the. Conclusively, knockdown of ZEB1 suppressed embryo implantation in vitro, paralleled with alteration of EMT markers. ZEB1 is likely to modulate endometrial receptivity through promotion of EMT, that could be crucial for embryo implantation process.
Collapse
Affiliation(s)
- Jing Ran
- The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, PR China
| | - Huan-Huan Yang
- The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, PR China
| | - Hui-Ping Huang
- The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, PR China
| | - Hong-Lang Huang
- The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, PR China
| | - Zhong Xu
- The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, PR China
| | - Wei Zhang
- The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, PR China; Department of Basic Medicine, School of Medicine, Xiamen University, Xiamen, 361102, PR China.
| | - Zhan-Xiang Wang
- The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, PR China.
| |
Collapse
|
37
|
Ion Channels in The Pathogenesis of Endometriosis: A Cutting-Edge Point of View. Int J Mol Sci 2020; 21:ijms21031114. [PMID: 32046116 PMCID: PMC7037987 DOI: 10.3390/ijms21031114] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/24/2020] [Accepted: 02/05/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Ion channels play a crucial role in many physiological processes. Several subtypes are expressed in the endometrium. Endometriosis is strictly correlated to estrogens and it is evident that expression and functionality of different ion channels are estrogen-dependent, fluctuating between the menstrual phases. However, their relationship with endometriosis is still unclear. OBJECTIVE To summarize the available literature data about the role of ion channels in the etiopathogenesis of endometriosis. METHODS A search on PubMed and Medline databases was performed from inception to November 2019. RESULTS Cystic fibrosis transmembrane conductance regulator (CFTR), transient receptor potentials (TRPs), aquaporins (AQPs), and chloride channel (ClC)-3 expression and activity were analyzed. CFTR expression changed during the menstrual phases and was enhanced in endometriosis samples; its overexpression promoted endometrial cell proliferation, migration, and invasion throughout nuclear factor kappa-light-chain-enhancer of activated B cells-urokinase plasminogen activator receptor (NFκB-uPAR) signaling pathway. No connection between TRPs and the pathogenesis of endometriosis was found. AQP5 activity was estrogen-increased and, through phosphatidylinositol-3-kinase and protein kinase B (PI3K/AKT), helped in vivo implantation of ectopic endometrium. In vitro, AQP9 participated in extracellular signal-regulated kinases/p38 mitogen-activated protein kinase (ERK/p38 MAPK) pathway and helped migration and invasion stimulating matrix metalloproteinase (MMP)2 and MMP9. ClC-3 was also overexpressed in ectopic endometrium and upregulated MMP9. CONCLUSION Available evidence suggests a pivotal role of CFTR, AQPs, and ClC-3 in endometriosis etiopathogenesis. However, data obtained are not sufficient to establish a direct role of ion channels in the etiology of the disease. Further studies are needed to clarify this relationship.
Collapse
|
38
|
Jing R, Kong Y, Han G, Zhang J, Li K, Dong X, Yan J, Zhang H, Han J, Feng L. The Mutation of the Ap3b1 Gene Causes Uterine Hypoplasia in Pearl Mice. Reprod Sci 2020; 27:182-191. [DOI: 10.1007/s43032-019-00006-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 03/25/2019] [Indexed: 11/30/2022]
|
39
|
Zeng S, Ulbrich SE, Bauersachs S. Spatial organization of endometrial gene expression at the onset of embryo attachment in pigs. BMC Genomics 2019; 20:895. [PMID: 31752681 PMCID: PMC6873571 DOI: 10.1186/s12864-019-6264-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 11/05/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND During the preimplantation phase in the pig, the conceptus trophoblast elongates into a filamentous form and secretes estrogens, interleukin 1 beta 2, interferons, and other signaling molecules before attaching to the uterine epithelium. The processes in the uterine endometrium in response to conceptus signaling are complex. Thus, the objective of this study was to characterize transcriptome changes in porcine endometrium during the time of conceptus attachment considering the specific localization in different endometrial cell types. RESULTS Low-input RNA-sequencing was conducted for the main endometrial compartments, luminal epithelium (LE), glandular epithelium (GE), blood vessels (BV), and stroma. Samples were isolated from endometria collected on Day 14 of pregnancy and the estrous cycle (each group n = 4) by laser capture microdissection. The expression of 12,000, 11,903, 11,094, and 11,933 genes was detectable in LE, GE, BV, and stroma, respectively. Differential expression analysis was performed between the pregnant and cyclic group for each cell type as well as for a corresponding dataset for complete endometrium tissue samples. The highest number of differentially expressed genes (DEGs) was found for LE (1410) compared to GE, BV, and stroma (800, 1216, and 384). For the complete tissue, 3262 DEGs were obtained. The DEGs were assigned to Gene Ontology (GO) terms to find overrepresented functional categories and pathways specific for the individual endometrial compartments. GO classification revealed that DEGs in LE were involved in 'biosynthetic processes', 'related to ion transport', and 'apoptotic processes', whereas 'cell migration', 'cell growth', 'signaling', and 'metabolic/biosynthetic processes' categories were enriched for GE. For blood vessels, categories such as 'focal adhesion', 'actin cytoskeleton', 'cell junction', 'cell differentiation and development' were found as overrepresented, while for stromal samples, most DEGs were assigned to 'extracellular matrix', 'gap junction', and 'ER to Golgi vesicles'. CONCLUSIONS The localization of differential gene expression to different endometrial cell types provided a significantly improved view on the regulation of biological processes involved in conceptus implantation, such as the control of uterine fluid secretion, trophoblast attachment, growth regulation by Wnt signaling and other signaling pathways, as well as the modulation of the maternal immune system.
Collapse
Affiliation(s)
- Shuqin Zeng
- Genetics and Functional Genomics, Clinic of Reproductive Medicine, Department for Farm Animals, Vetsuisse Faculty, University of Zurich, Eschikon 27 AgroVet-Strickhof, Zurich, Switzerland
- Animal Physiology, Institute of Agricultural Sciences, ETH Zurich, Lindau, ZH 8315 Switzerland
| | - Susanne E. Ulbrich
- Animal Physiology, Institute of Agricultural Sciences, ETH Zurich, Lindau, ZH 8315 Switzerland
| | - Stefan Bauersachs
- Genetics and Functional Genomics, Clinic of Reproductive Medicine, Department for Farm Animals, Vetsuisse Faculty, University of Zurich, Eschikon 27 AgroVet-Strickhof, Zurich, Switzerland
| |
Collapse
|
40
|
Fatima SS, Rehman R, Martins RS, Alam F, Ashraf M. Single nucleotide polymorphisms in Renalase and KCNQ1 genes and female infertility: A cross-sectional study in Pakistan. Andrologia 2019; 51:e13434. [PMID: 31579970 DOI: 10.1111/and.13434] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/19/2019] [Accepted: 08/27/2019] [Indexed: 01/21/2023] Open
Abstract
A global increase in the incidence of subfertility is observed, and research suggests strong genetic influences that might restrict fertility directly or indirectly. It therefore becomes important to rule out the existence of genetic causes and counsel infertile couples before offering "Advanced Infertility Treatment Techniques." This cross-sectional study aimed to explore the association of KCNQ1 (rs2237895) and Renalase (rs2576178 and rs10887800) single nucleotide polymorphisms with different causes of infertility by analysing 508 fertile and 164 infertile women. Gene variant (AC/CC) of KCNQ1 rs2237895 showed a slight difference in the endometriosis group compared to the fertile group (p = .049), with the C allele showing a significant association with infertility overall (OR = 1.42 [1.100-1.833]; p < .0069). The variant AG/GG of Renalase rs2576178 was significantly associated with overall infertility (OR = 2.266; p < .001), with a strong G allele association with unexplained infertility OR = 2.796 (p = .002) that remained significant after adjusting for age and body mass index. Similarly, Renalase rs10887800 AG/GG and G allele showed significant association with both infertility due to polycystic ovarian syndrome and unexplained infertility. Expression of single nucleotide polymorphism rs2237895 and rs2576178 in both KCNQ1 and Renalase genes might be responsible for altering reproductive potential, hence leading to infertility in women.
Collapse
Affiliation(s)
- Syeda Sadia Fatima
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | - Rehana Rehman
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | | | - Faiza Alam
- Department of Physiology, University of Karachi, Karachi, Pakistan
| | - Mussarat Ashraf
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| |
Collapse
|
41
|
Wetendorf M, Randall LT, Lemma MT, Hurr SH, Pawlak JB, Tarran R, Doerschuk CM, Caron KM. E-Cigarette Exposure Delays Implantation and Causes Reduced Weight Gain in Female Offspring Exposed In Utero. J Endocr Soc 2019; 3:1907-1916. [PMID: 31598571 PMCID: PMC6777403 DOI: 10.1210/js.2019-00216] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 08/15/2019] [Indexed: 12/25/2022] Open
Abstract
Electronic nicotine delivery system (e-cigarette) use is prevalent among pregnant women as a seemingly safe alternative to traditional tobacco use, known to result in fetal developmental abnormalities and impaired fertility of male offspring. However, little is known about the effects of e-cigarette use on fertility or pregnancy outcomes. A successful pregnancy is initiated by a multitude of dynamic molecular alterations in the uterus resulting in embryo implantation at day 4.5 in the mouse. We examined whether e-cigarette exposure impairs implantation and offspring health. Pregnant C57BL/6J mice were exposed five times a week to e-cigarette vapor or sham. After 4 months, e-cigarette exposed dams exhibited a significant delay in the onset of the first litter. Furthermore, exposure of new dams in early pregnancy significantly impaired embryo implantation, as evidenced by nearly complete absence of implantation sites in e-cigarette-exposed animals at day 5.5, despite exhibiting high levels of progesterone, an indicator of pregnancy. RNA microarray from day 4.5 pseudopregnant mice revealed significant changes in the integrin, chemokine, and JAK signaling pathways. Moreover, female offspring exposed to e-cigarettes in utero exhibited a significant weight reduction at 8.5 months, whereas males exhibited a slight but nonsignificant deficiency in fertility. Thus, e-cigarette exposure in mice impairs pregnancy initiation and fetal health, suggesting that e-cigarette use by reproductive-aged women or during pregnancy should be considered with caution.
Collapse
Affiliation(s)
- Margeaux Wetendorf
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina
| | - Lewis T Randall
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Mahlet T Lemma
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Sophia H Hurr
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina
| | - John B Pawlak
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina
| | - Robert Tarran
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina
| | - Claire M Doerschuk
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
42
|
Xu JW, Gao DD, Peng L, Qiu ZE, Ke LJ, Zhu YX, Zhang YL, Zhou WL. The gasotransmitter hydrogen sulfide inhibits transepithelial anion secretion of pregnant mouse endometrial epithelium. Nitric Oxide 2019; 90:37-46. [PMID: 31175932 DOI: 10.1016/j.niox.2019.05.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 05/24/2019] [Accepted: 05/29/2019] [Indexed: 01/08/2023]
Abstract
Endometrial epithelium exhibits a robust ion transport activity required for dynamical regulation of uterine fluid environment and thus embryo implantation. However, there still lacks a thorough understanding of the ion transport processes and regulatory mechanism in peri-implantation endometrial epithelium. As a gaseous signaling molecule or gasotransmitter, hydrogen sulfide (H2S) regulates a myriad of cellular and physiological processes in various tissues, including the modulation of ion transport proteins in epithelium. This study aimed to investigate the effects of H2S on ion transport across mouse endometrial epithelium and its possible role in embryo implantation. The existence of endogenous H2S in pregnant mouse uterus was tested by the detection of two key H2S-generating enzymes and measurement of H2S production rate in tissue homogenates. Transepithelial ion transport processes were electrophysiologically assessed in Ussing chambers on early pregnant mouse endometrial epithelial layers, demonstrating that H2S suppressed the anion secretion by blocking cystic fibrosis transmembrane conductance regulator (CFTR). H2S increased intracellular Cl- concentration ([Cl-]i) in mouse endometrial epithelial cells, which was abolished by pretreatment with the CFTR selective inhibitor CFTRinh-172. The cAMP level in mouse endometrial epithelial cells was not affected by H2S, indicating that H2S blocked CFTR in a cAMP-independent way. In vivo study showed that interference with H2S synthesis impaired embryo implantation. In conclusion, our study demonstrated that H2S inhibits the transepithelial anion secretion of early pregnant mouse endometrial epithelium via blockade of CFTR, contributing to the preparation for embryo implantation.
Collapse
Affiliation(s)
- Jia-Wen Xu
- School of Life Sciences, Sun Yat-sen University, 510006, No. 132, Waihuan Dong Road, Higher Education Mega Center, Guangzhou, China
| | - Dong-Dong Gao
- School of Life Sciences, Sun Yat-sen University, 510006, No. 132, Waihuan Dong Road, Higher Education Mega Center, Guangzhou, China
| | - Lei Peng
- School of Life Sciences, Sun Yat-sen University, 510006, No. 132, Waihuan Dong Road, Higher Education Mega Center, Guangzhou, China
| | - Zhuo-Er Qiu
- School of Life Sciences, Sun Yat-sen University, 510006, No. 132, Waihuan Dong Road, Higher Education Mega Center, Guangzhou, China
| | - Li-Jiao Ke
- School of Life Sciences, Sun Yat-sen University, 510006, No. 132, Waihuan Dong Road, Higher Education Mega Center, Guangzhou, China
| | - Yun-Xin Zhu
- School of Life Sciences, Sun Yat-sen University, 510006, No. 132, Waihuan Dong Road, Higher Education Mega Center, Guangzhou, China
| | - Yi-Lin Zhang
- School of Life Sciences, Sun Yat-sen University, 510006, No. 132, Waihuan Dong Road, Higher Education Mega Center, Guangzhou, China.
| | - Wen-Liang Zhou
- School of Life Sciences, Sun Yat-sen University, 510006, No. 132, Waihuan Dong Road, Higher Education Mega Center, Guangzhou, China.
| |
Collapse
|
43
|
A High Protein Model Alters the Endometrial Transcriptome of Mares. Genes (Basel) 2019; 10:genes10080576. [PMID: 31366166 PMCID: PMC6723232 DOI: 10.3390/genes10080576] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/26/2019] [Accepted: 07/22/2019] [Indexed: 01/11/2023] Open
Abstract
High blood urea nitrogen (BUN) decreases fertility of several mammals; however, the mechanisms have not been investigated in mares. We developed an experimental model to elevate BUN, with urea and control treatments (7 mares/treatment), in a crossover design. Urea-treatment consisted of a loading dose of urea (0.03 g/kg of body weight (BW)) and urea injections over 6 hours (0.03 g/kg of BW/h). Control mares received the same volume of saline solution. Blood samples were collected to measure BUN. Uterine and vaginal pH were evaluated after the last intravenous infusion, then endometrial biopsies were collected for RNA-sequencing with a HiSeq 4000. Cuffdiff (2.2.1) was used to identify the differentially expressed genes (DEG) between urea and control groups (false discovery rate-adjusted p-value < 0.1). There was a significant increase in BUN and a decrease of uterine pH in the urea group compared to the control group. A total of 193 genes were DEG between the urea and control groups, with five genes identified as upstream regulators (ETV4, EGF, EHF, IRS2, and SGK1). The DEG were predicted to be related to cell pH, ion homeostasis, changes in epithelial tissue, and solute carriers. Changes in gene expression reveal alterations in endometrial function that could be associated with adverse effects on fertility of mares.
Collapse
|
44
|
Sohn JO, Seong SY, Kim HJ, Jo YM, Lee KH, Chung MK, Song HJ, Park KS, Lim JM. Alterations in intracellular Ca 2+ levels in human endometrial stromal cells after decidualization. Biochem Biophys Res Commun 2019; 515:318-324. [PMID: 31153638 DOI: 10.1016/j.bbrc.2019.05.153] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 05/24/2019] [Indexed: 01/17/2023]
Abstract
Calcium (Ca2+) is an important element for many physiological functions of the uterus, including embryo implantation. Here, we investigated the possible involvement of altered intracellular Ca2+ levels in decidualization in human endometrial stromal cells (hEMSCs). hEMSCs showed high levels of mesenchymal stem cell marker expression (CD73, CD90, and CD105) and did not express markers of hematopoietic progenitor cells (CD31, CD34, CD45, and HLA-DR). Decidualization is a process of ovarian steroid-induced endometrial stromal cell proliferation and differentiation. Several types of ion channels, which are regulated by the ovarian hormones progesterone and estradiol, as well as growth factors, are important for endometrial receptivity and embryo implantation. The combined application of progesterone (1 μM medroxyprogesterone acetate) and cyclic AMP (0.5 mM) for 6 days not only elevated inositol 1,4,5-triphosphate receptor (IP3R)-mediated Ca2+ release and IP3R expression, it also promoted ORAI and STIM expression as well as cyclopiazonic acid-induced Ca2+ release. Finally, intracellular Ca2+ levels and ion channel gene expression influenced hEMSC proliferation. These results suggest that cytosolic Ca2+ dynamics, mediated by specific ion channels, serve as an important step in the decidualization of hEMSCs.
Collapse
Affiliation(s)
- Jie Ohn Sohn
- Department of Agricultural Biotechnology, Seoul National University, Seoul, 151-921, South Korea; Fertility Medical Center, Seoul Women's Hospital, Bucheon, 14544, South Korea
| | - Seung Yong Seong
- Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, 25159, South Korea
| | - Hyun Jin Kim
- Department of Physiology, Sungkyunkwan University School of Medicine, Suwon, 16419, South Korea
| | - Yoon Mi Jo
- Fertility Medical Center, Seoul Women's Hospital, Bucheon, 14544, South Korea
| | - Kyoung Hoon Lee
- Fertility Medical Center, Seoul Women's Hospital, Bucheon, 14544, South Korea
| | - Mi Kyung Chung
- Seoul Rachel Fertility Center, Seoul, 04146, South Korea
| | - Hyun Jin Song
- Fertility Medical Center, Seoul Women's Hospital, Bucheon, 14544, South Korea
| | - Kyoung Sun Park
- Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon, 25159, South Korea.
| | - Jeong Mook Lim
- Department of Agricultural Biotechnology, Seoul National University, Seoul, 151-921, South Korea; Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 151-921, South Korea.
| |
Collapse
|
45
|
Chen JJ, Xiao ZJ, Meng X, Wang Y, Yu MK, Huang WQ, Sun X, Chen H, Duan YG, Jiang X, Wong MP, Chan HC, Zou F, Ruan YC. MRP4 sustains Wnt/β-catenin signaling for pregnancy, endometriosis and endometrial cancer. Am J Cancer Res 2019; 9:5049-5064. [PMID: 31410201 PMCID: PMC6691374 DOI: 10.7150/thno.32097] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 05/08/2019] [Indexed: 12/13/2022] Open
Abstract
Rationale: Abnormal Wnt/β-catenin signaling in the endometrium can lead to both embryo implantation failure and severe pathogenic changes of the endometrium such as endometrial cancer and endometriosis. However, how Wnt/β-catenin signaling is regulated in the endometrium remains elusive. We explored possible regulation of Wnt/β-catenin signaling by multi-drug resistance protein 4 (MRP4), a potential target in cancer chemotherapy, and investigated the mechanism. Methods: Knockdown of MRP4 was performed in human endometrial cells in vitro or in a mouse embryo-implantation model in vivo. Immunoprecipitation, immunoblotting and immunofluorescence were used to assess protein interaction and stability. Wnt/β-catenin signaling was assessed by TOPflash reporter assay and quantitative PCR array. Normal and endometriotic human endometrial tissues were examined. Data from human microarray or RNAseq databases of more than 100 participants with endometriosis, endometrial cancer or IVF were analyzed. In vitro and in vivo tumorigenesis was performed. Results: MRP4-knockdown, but not its transporter-function-inhibition, accelerates β-catenin degradation in human endometrial cells. MRP4 and β-catenin are co-localized and co-immunoprecipitated in mouse and human endometrium. MRP4-knockdown in mouse uterus reduces β-catenin levels, downregulates a series of Wnt/β-catenin target genes and impairs embryo implantation, which are all reversed by blocking β-catenin degradation. Analysis of human endometrial biopsy samples and available databases reveals significant and positive correlations of MRP4 with β-catenin and Wnt/β-catenin target genes in the receptive endometrium in IVF, ectopic endometriotic lesions and endometrial cancers. Knockdown of MRP4 also inhibits in vitro and in vivo endometrial tumorigenesis. Conclusion: A previously undefined role of MRP4 in stabilizing β-catenin to sustain Wnt/β-catenin signaling in endometrial cells is revealed for both embryo implantation and endometrial disorders, suggesting MRP4 as a theranostic target for endometrial diseases associated with Wnt/β-catenin signaling abnormality.
Collapse
|
46
|
Ghavideldarestani M, Butler AE, Shirian S, Atkin SL. Expression and localization of transient receptor potential channels in the bovine uterus epithelium throughout the estrous cycle. Mol Biol Rep 2019; 46:4077-4084. [PMID: 31087243 DOI: 10.1007/s11033-019-04857-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 05/03/2019] [Indexed: 12/14/2022]
Abstract
Transient receptor potential (TRP) channels are expressed in the endometrium but it is unknown if they are modulated through the estrous cycle (EC). This study was undertaken to identify the modulation of the TRPC gene and protein isoforms in bovine uterine epithelium, as a model for human, throughout the EC. Changes in the expression of TRPC genes in bovine uterine epithelium throughout the EC were measured using Real-Time PCR, while immunohistochemistry and immunocytochemistry were used to determine the localization of these channels. Out of the 7 members of the TRPC family, TRPC1, 2, 3, 4 and 6 genes were expressed in bovine uterine epithelial tissue and TRPC 5 and 7 were not. Gene expression levels of all TRPC isoforms underwent cyclical changes throughout the EC. Moreover, cyclical changes were detected in the protein levels of TRPC1 and TRPC6 throughout the EC. These findings show that TRPC channels are modulated through the EC and therefore may have a role in reproductive events.
Collapse
Affiliation(s)
| | | | - Sadegh Shirian
- Department of Pathology, School of Veterinary Medicine, Shahrekord University, Shahrekord, Iran.,Shiraz Molecular Pathology Research Center, Dr Daneshbod Pathol Lab, Shiraz, Iran.,Biotechnology Research Inistitute, Shahrekord University, Shahrekord, Iran
| | - Stephen L Atkin
- Weill Cornell Medical College Qatar, Qatar Foundation, Education City, PO Box 24144, Doha, Qatar.
| |
Collapse
|
47
|
Hennes A, Held K, Boretto M, De Clercq K, Van den Eynde C, Vanhie A, Van Ranst N, Benoit M, Luyten C, Peeraer K, Tomassetti C, Meuleman C, Voets T, Vankelecom H, Vriens J. Functional expression of the mechanosensitive PIEZO1 channel in primary endometrial epithelial cells and endometrial organoids. Sci Rep 2019; 9:1779. [PMID: 30741991 PMCID: PMC6370865 DOI: 10.1038/s41598-018-38376-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 12/20/2018] [Indexed: 12/27/2022] Open
Abstract
Successful pregnancy requires the establishment of a complex dialogue between the implanting embryo and the endometrium. Knowledge regarding molecular candidates involved in this early communication process is inadequate due to limited access to primary human endometrial epithelial cells (EEC). Since pseudo-pregnancy in rodents can be induced by mechanical scratching of an appropriately primed uterus, this study aimed to investigate the expression of mechanosensitive ion channels in EEC. Poking of EEC provoked a robust calcium influx and induced an increase in current densities, which could be blocked by an inhibitor of mechanosensitive ion channels. Interestingly, RNA expression studies showed high expression of PIEZO1 in EEC of mouse and human. Additional analysis provided further evidence for the functional expression of PIEZO1 since stimulation with Yoda1, a chemical agonist of PIEZO1, induced increases in intracellular calcium concentrations and current densities in EEC. Moreover, the ion channel profile of human endometrial organoids (EMO) was validated as a representative model for endometrial epithelial cells. Mechanical and chemical stimulation of EMO induced strong calcium responses supporting the hypothesis of mechanosensitive ion channel expression in endometrial epithelial cells. In conclusion, EEC and EMO functionally express the mechanosensitive PIEZO1 channel that could act as a potential target for the development of novel treatments to further improve successful implantation processes.
Collapse
Affiliation(s)
- Aurélie Hennes
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Herestraat 49 box 611, 3000, Leuven, Belgium
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, VIB Center for Brain & Disease Research, Herestraat 49 box 802, 3000, Leuven, Belgium
| | - Katharina Held
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Herestraat 49 box 611, 3000, Leuven, Belgium
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, VIB Center for Brain & Disease Research, Herestraat 49 box 802, 3000, Leuven, Belgium
| | - Matteo Boretto
- Laboratory of Tissue Plasticity in Health and Disease, Cluster of Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven, Herestraat 49 box 804, 3000, Leuven, Belgium
| | - Katrien De Clercq
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Herestraat 49 box 611, 3000, Leuven, Belgium
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, VIB Center for Brain & Disease Research, Herestraat 49 box 802, 3000, Leuven, Belgium
| | - Charlotte Van den Eynde
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Herestraat 49 box 611, 3000, Leuven, Belgium
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, VIB Center for Brain & Disease Research, Herestraat 49 box 802, 3000, Leuven, Belgium
| | - Arne Vanhie
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Herestraat 49 box 611, 3000, Leuven, Belgium
- Leuven University Fertility Centre, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Nele Van Ranst
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, VIB Center for Brain & Disease Research, Herestraat 49 box 802, 3000, Leuven, Belgium
| | - Melissa Benoit
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, VIB Center for Brain & Disease Research, Herestraat 49 box 802, 3000, Leuven, Belgium
| | - Catherine Luyten
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Herestraat 49 box 611, 3000, Leuven, Belgium
| | - Karen Peeraer
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Herestraat 49 box 611, 3000, Leuven, Belgium
- Leuven University Fertility Centre, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Carla Tomassetti
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Herestraat 49 box 611, 3000, Leuven, Belgium
- Leuven University Fertility Centre, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Christel Meuleman
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Herestraat 49 box 611, 3000, Leuven, Belgium
- Leuven University Fertility Centre, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Thomas Voets
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, VIB Center for Brain & Disease Research, Herestraat 49 box 802, 3000, Leuven, Belgium
| | - Hugo Vankelecom
- Laboratory of Tissue Plasticity in Health and Disease, Cluster of Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven, Herestraat 49 box 804, 3000, Leuven, Belgium
| | - Joris Vriens
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, KU Leuven, Herestraat 49 box 611, 3000, Leuven, Belgium.
| |
Collapse
|
48
|
Wang X, Yu Q. Endometriosis-related ceRNA network to identify predictive biomarkers of endometrial receptivity. Epigenomics 2019; 11:147-167. [PMID: 30638056 DOI: 10.2217/epi-2018-0190] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIM As RNA, which plays a role in the regulation of endometrial receptivity, can be modulated via ceRNA mechanisms, we constructed a ceRNA network to explore potential RNA/ceRNA biomarkers indicating endometrial receptivity associated with endometriosis. MATERIALS & METHODS RNA sequencing was performed on eutopic endometrium from eight patients with and without endometriosis. Bioinformatics algorithms were used to predict ceRNA network and pathway analysis. RESULTS We identified an endometriosis-associated ceRNA network involving 45 pathways and four ceRNAs as potential predictive biomarkers for endometrial receptivity. Patients with endometriosis presented lower levels of progesterone receptor type B expression. CONCLUSION Differentially expressed RNAs and lower progesterone receptors type B levels in endometriosis might be related to the impairment of endometrial receptivity.
Collapse
Affiliation(s)
- Xi Wang
- Department of Obstetrics & Gynaecology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| | - Qi Yu
- Department of Obstetrics & Gynaecology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
49
|
A Two-Cohort RNA-seq Study Reveals Changes in Endometrial and Blood miRNome in Fertile and Infertile Women. Genes (Basel) 2018; 9:genes9120574. [PMID: 30477193 PMCID: PMC6315937 DOI: 10.3390/genes9120574] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/07/2018] [Accepted: 11/07/2018] [Indexed: 12/16/2022] Open
Abstract
The endometrium undergoes extensive changes to prepare for embryo implantation and microRNAs (miRNAs) have been described as playing a significant role in the regulation of endometrial receptivity. However, there is no consensus about the miRNAs involved in mid-secretory endometrial functions. We analysed the complete endometrial miRNome from early secretory (pre-receptive) and mid-secretory (receptive) phases from fertile women and from patients with recurrent implantation failure (RIF) to reveal differentially expressed (DE) miRNAs in the mid-secretory endometrium. Furthermore, we investigated whether the overall changes during early to mid-secretory phase transition and with RIF condition could be reflected in blood miRNA profiles. In total, 116 endometrial and 114 matched blood samples collected from two different population cohorts were subjected to small RNA sequencing. Among fertile women, 91 DE miRNAs were identified in the mid-secretory vs. early secretory endometrium, while no differences were found in the corresponding blood samples. The comparison of mid-secretory phase samples between fertile and infertile women revealed 21 DE miRNAs from the endometrium and one from blood samples. Among discovered novel miRNAs, chr2_4401 was validated and showed up-regulation in the mid-secretory endometrium. Besides novel findings, we confirmed the involvement of miR-30 and miR-200 family members in mid-secretory endometrial functions.
Collapse
|
50
|
Boggula VR, Hanukoglu I, Sagiv R, Enuka Y, Hanukoglu A. Expression of the epithelial sodium channel (ENaC) in the endometrium - Implications for fertility in a patient with pseudohypoaldosteronism. J Steroid Biochem Mol Biol 2018; 183:137-141. [PMID: 29885352 DOI: 10.1016/j.jsbmb.2018.06.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 06/01/2018] [Accepted: 06/05/2018] [Indexed: 01/21/2023]
Abstract
Pseudohypoaldosteronism type 1 (PHA) is a syndrome of unresponsiveness to aldosterone. The severe form of this disease results from mutations in the genes that encode for the epithelial sodium channel subunits, SCNN1A, SCNN1B, and SCNN1G. A PHA patient under our care failed to conceive after many years and IVF trials. Our earlier studies had shown that ENaC is expressed in the female reproductive tract. We hypothesized that a defective ENaC expression may be responsible for the infertility of the patient. To test this hypothesis we examined ENaC expression in endometrial Pipelle biopsy samples from three healthy women and the PHA patient with an Arg508X mutation in the SCNN1A gene. The formalin fixed samples were reacted with anti-ENaCA (alpha subunit) antisera, followed by secondary antibodies to visualize ENaC expression by immunofluorescence. Confocal microscopy imaging of the samples showed strong ENaC immunofluorescence along the luminal border (apical membrane) of the epithelial cells in Pipelle samples from healthy women. In contrast, none of the samples from the PHA patient showed ENaC immunofluorescence. The Arg508X mutation interrupts the transport of ENaC subunits to the cell surface, yet it would not be expected to disrupt ENaC localization in the cytoplasm. In contrast to endometrium where ENaC is localized in the apical membrane of the epithelial cells, in keratinocytes ENaC is expressed in cytoplasmic pools. Therefore, we examined ENaC immunofluorescence in plucked hair follicles. As expected, ENaC immunofluorescence was detected in the cytoplasm of keratinocytes of both normal and PHA samples. Our results support the hypothesis that lack of expression of ENaC on the endometrial surface may be responsible for the infertility of the PHA patient.
Collapse
Affiliation(s)
| | | | - Ron Sagiv
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel; Department of Obstetrics & Gynecology, E. Wolfson Medical Center, Holon, Israel
| | - Yehoshua Enuka
- Laboratory of Cell Biology, Ariel University, Ariel, Israel
| | - Aaron Hanukoglu
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel; Division of Pediatric Endocrinology, E. Wolfson Medical Center, Holon, Israel.
| |
Collapse
|