1
|
Fujimura T, Tamura I, Yoshimura A, Yoneda T, Takasaki H, Shiroshita A, Shirafuta Y, Sato S, Sugino N. Establishment of an in vitro implantation model using a newly developed mouse endometrial organoid. Development 2025; 152:dev204461. [PMID: 40365775 PMCID: PMC12091871 DOI: 10.1242/dev.204461] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 03/17/2025] [Indexed: 05/15/2025]
Abstract
Implantation failure is a major cause of infertility, but its mechanisms remain unclear due to the lack of techniques for constructing organized endometrial structures and recapitulating the implantation process in vitro. Endometrial organoids have recently been developed, but they consist of only epithelial cells, and their apical surface faces inward, preventing blastocyst attachment. We developed an apical-out mouse endometrial organoid incorporating epithelial and stromal cells, and examined its ability to recapitulate implantation with mouse blastocysts. Mouse uteri were digested with collagenase and cultured in monolayers. The resulting aggregates were then transferred to low-attachment plates for 3D culture. After 7 days, self-organized aggregates contained E-cadherin-positive epithelial cells outside and vimentin-positive stromal cells inside. Mucin 1 signals were observed on the apical side of epithelial cells, confirming the apical-out orientation. Organoids were stimulated with sex steroid hormones and co-cultured with blastocysts. Time-lapse imaging revealed the four implantation steps: blastocyst attachment, epithelial invagination, entosis and invasion. Invaded cells expressed proliferin while surrounding stromal cells expressed cyclooxygenase 2, indicating trophoblast differentiation and decidualization. This novel organoid closely recapitulates the mouse endometrium and implantation process in vitro.
Collapse
Affiliation(s)
- Taishi Fujimura
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Minamikogushi 1-1-1, Ube 755-8505, Japan
| | - Isao Tamura
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Minamikogushi 1-1-1, Ube 755-8505, Japan
| | - Azumi Yoshimura
- Yamaguchi University Graduate School of Medicine, Minamikogushi 1-1-1, Ube 755-8505, Japan
| | - Toshihide Yoneda
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Minamikogushi 1-1-1, Ube 755-8505, Japan
| | - Hitomi Takasaki
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Minamikogushi 1-1-1, Ube 755-8505, Japan
| | - Amon Shiroshita
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Minamikogushi 1-1-1, Ube 755-8505, Japan
| | - Yuichiro Shirafuta
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Minamikogushi 1-1-1, Ube 755-8505, Japan
| | - Shun Sato
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Minamikogushi 1-1-1, Ube 755-8505, Japan
| | - Norihiro Sugino
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Minamikogushi 1-1-1, Ube 755-8505, Japan
| |
Collapse
|
2
|
Yang Y, Du Y, Ma X, Yuan G, Li G, Zhang Q, Zhou S. Transcription factor addictions: exploring the potential Achilles' Heel of endometriosis. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-024-2832-8. [PMID: 40163264 DOI: 10.1007/s11427-024-2832-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 11/15/2024] [Indexed: 04/02/2025]
Abstract
A considerable number of women of reproductive age suffer from endometriosis worldwide. There is a significant physical, mental, and financial burden on patients affected by this condition in terms of pelvic pain, either continuously or intermittently, dysmenorrhea, infertility, and a higher risk of certain types of cancer. Several treatments available in clinical settings for endometriosis management do not provide adequate efficacy and have undesirable side effects. Transcription factors (TFs) are crucial regulators of key biological processes involved in endometriosis. Here, we elaborated on the research progress regarding the crucial roles of TFs in endometriosis, emphasizing their implications for clinical outcomes and critical therapeutic contributions. By delving into their involvement in key processes, such as cell proliferation and apoptosis, we revealed the multifaceted role of key TFs in disease progression. We aimed to provide a systemic understanding of TFs regulation in endometriosis pathogenesis, establishing a foundation for innovative treatment approaches.
Collapse
Affiliation(s)
- Yang Yang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China
| | - Yi Du
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China
| | - Xuelei Ma
- Department of Biotherapy, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Gang Yuan
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Guobo Li
- Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Qian Zhang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China.
| | - Shengtao Zhou
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, 610041, China.
| |
Collapse
|
3
|
He W, Loganathan N, Belsham DD. IGF1 Signaling Regulates Neuropeptide Expression in Hypothalamic Neurons Under Physiological and Pathological Conditions. Endocrinology 2025; 166:bqaf051. [PMID: 40105689 PMCID: PMC11949690 DOI: 10.1210/endocr/bqaf051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/28/2025] [Accepted: 03/17/2025] [Indexed: 03/20/2025]
Abstract
Insulin-like growth factor 1 (IGF1) plays a critical role in metabolism and aging, but its role in the brain remains unclear. This study examined whether hypothalamic neurons respond to IGF1 and how its actions are modulated. RT-qPCR and single-cell RNA sequencing indicated that Igf1r mRNA is expressed in neuropeptide Y/Agouti-related peptide (NPY/AgRP) neurons but has higher expression in pro-opiomelanocortin (POMC) neurons. IGF1 binding proteins Igfbp3 and Igfbp5 were significantly expressed, whereby Igfbp5 levels were modulated by fasting, nutrient availability, and circadian rhythms, implying that IGF1 signaling can be controlled by multiple mechanisms. In mouse and human models, IGF1 regulated Agrp, Npy, Pomc, Cartpt, Spx, Gal, and Fam237b expression, producing an overall anorexigenic profile. Hyperinsulinemia induced IGF1 resistance, accompanied by reduced IGF1R protein, as well as Igf1r and Irs2 mRNA expression via over-activation of phosphoinositide 3-kinase/forkhead box O1 (PI3K-FOXO1) signaling. Thus, hypothalamic neurons respond to IGF1 under physiological conditions, and hyperinsulinemia is a novel mechanism that drives cellular IGF1 resistance.
Collapse
Affiliation(s)
- Wenyuan He
- Department of Physiology, University of Toronto, Toronto, ON, Canada M5S 1A8
| | - Neruja Loganathan
- Department of Physiology, University of Toronto, Toronto, ON, Canada M5S 1A8
| | - Denise D Belsham
- Department of Physiology, University of Toronto, Toronto, ON, Canada M5S 1A8
- Department of Medicine, University of Toronto, Toronto, ON, Canada M5S 1A8
- Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON, Canada M5S 1A8
| |
Collapse
|
4
|
Jia Z, Li B, Matsuo M, Dewar A, Mustafaraj A, Dey SK, Yuan J, Sun X. Foxa2-dependent uterine glandular cell differentiation is essential for successful implantation. Nat Commun 2025; 16:2465. [PMID: 40074766 PMCID: PMC11904179 DOI: 10.1038/s41467-025-57848-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
Uterine receptivity is essential for successful implantation. In mice, uterine receptivity begins with the secretion of LIF from uterine glands stimulated by estrogen on the morning of day 4 pregnancy. We hypothesize that gland readiness for estrogen stimulation is indispensable for uterine receptivity. The current study reveals that uterine glands undergo a differentiation process with expanded branching during the preimplantation period. The single-cell RNA profiling of glandular cells identifies that LIF is expressed exclusively in a Prss29+ subgroup of glandular cells on day 4 of pregnancy. Interestingly, Foxa2-deficient glands lacking LIF production fail to develop branches and the functional Prss29+ subgroup. This Prss29+ subgroup develops prior to estrogen secretion. Collectively, our findings show that uterine glands undergo a FOXA2-dependent maturation process to acquire the competence, named "transitional phase", for entering the receptive phase. The "transitional phase", predicting uterine receptivity one day before implantation, is a landmark concept in uterine receptivity.
Collapse
Affiliation(s)
- Zhaoyu Jia
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Bo Li
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Mitsunori Matsuo
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- College of Medicine, University of Cincinnati, Cincinnati, OH, USA
- Department of Obstetrics and Gynecology, University of Tokyo, Tokyo, Japan
| | - Amanda Dewar
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Anxhela Mustafaraj
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Sudhansu K Dey
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- College of Medicine, University of Cincinnati, Cincinnati, OH, USA.
| | - Jia Yuan
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| | - Xiaofei Sun
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- College of Medicine, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
5
|
Tran DN, Hwang YJ, Kim KC, Li R, Marquardt RM, Chen C, Young SL, Lessey BA, Kim TH, Cheon YP, Jeong JW. GRB2 regulation of essential signaling pathways in the endometrium is critical for implantation and decidualization. Nat Commun 2025; 16:2192. [PMID: 40038241 PMCID: PMC11880417 DOI: 10.1038/s41467-025-57173-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 02/12/2025] [Indexed: 03/06/2025] Open
Abstract
Over 75% of failed pregnancies involve implantation defects. Growth factor receptor-bound protein 2 (GRB2) is an adaptor protein involved in signal transduction and cell communication. Here we show that the expression of GRB2 protein is lower in endometrium of infertile women with endometriosis compared to controls. Our mouse endometriosis model revealed that endometriosis development results to GRB2 loss in the eutopic endometrium. To understand the role of GRB2 in the uterus, we generated mice with conditional ablation of Grb2 in the Pgr positive cells (Grb2d/d). Grb2d/d mice were infertile due to implantation failure. Although ovarian functions were normal, Grb2d/d mice had a non-receptive endometrium due to progesterone resistance and dysregulation of steroid hormone and FOXA2 signaling pathways. Furthermore, our results were supported by findings of GRB2 attenuation in primary human endometrial stromal cells from women with endometriosis. Our results demonstrate that GRB2 is critical for endometrial receptivity and decidualization.
Collapse
Affiliation(s)
- Dinh Nam Tran
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, Columbia, MO, USA
| | - Yeon Jeong Hwang
- Division of Developmental Biology and Physiology, Department of Biotechnology, Institute of Basic Sciences, Sungshin Women's University, Seoul, South Korea
| | - Keun Cheon Kim
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, Columbia, MO, USA
| | - Rong Li
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, Columbia, MO, USA
| | - Ryan M Marquardt
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, Grand Rapids, MI, USA
| | - Chen Chen
- Department of Animal Science, Michigan State University, East Lansing, MI, USA
| | - Steven L Young
- Department of Obstetrics, Gynecology and Women's Health, Duke University, Durham, NC, USA
| | - Bruce A Lessey
- Department of Obstetrics and Gynecology, Wake Forest Baptist Health, Winston-Salem, NC, USA
| | - Tae Hoon Kim
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, Columbia, MO, USA
| | - Yong-Pil Cheon
- Division of Developmental Biology and Physiology, Department of Biotechnology, Institute of Basic Sciences, Sungshin Women's University, Seoul, South Korea.
| | - Jae-Wook Jeong
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri School of Medicine, Columbia, MO, USA.
| |
Collapse
|
6
|
Xu S, Hu D, Ye Y, Mu Y, Xiong Y, Zhang Y. Identification of serum small non-coding RNA as biomarkers for endometrial receptivity. Genomics 2025; 117:111002. [PMID: 39848478 DOI: 10.1016/j.ygeno.2025.111002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 12/30/2024] [Accepted: 01/19/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Current endometrial receptivity analysis is invasive, preventing embryo transfer during the biopsy cycle. This study aims to screen serum sncRNAs as non-invasive biomarkers for ERA tests. METHODS The study included 12 infertile patients undergoing IVF-ET and ERA, whose serum samples were collected for high-energy sequencing technology to detect sncRNA expression profiles. We overexpressed and knocked down tsRNA-35:73-Asp-GTC-1 in the decidualized Immortalized Human Eutopic Endometrial Stromal Cells (HESC) model cultured in vitro to further investigate the its effect on decidualization. The predicted tsRNA-35:73-Asp-GTC-1 target gene was verified by PCR analysis. RESULTS We screened 286 differentially expressed tsRNAs, 46 miRNAs, and 106 piRNAs. KEGG analysis indicated that differentially expressed tsRNAs were associated with pathways such as 'Calcium signaling pathway,' 'Sphingolipid signaling pathway,' etc. The results of RT-qPCR validation showed that the trends of four significantly differentially expressed tsRNAs in serum and endometrium were consistent with sequencing results. ROC curves demonstrated that these four tsRNAs have good predictive value for endometrial receptivity. Overexpression of tsRNA-35:73-Asp-GTC-1 affected the morphology of decidualized cells, and the decidualization indicators also showed a decreasing trend. While knocking down tsRNA-35:73-Asp-GTC-1 had the opposite effect. The RT-qPCR results showed that tsRNA-35:73-Asp-GTC-1 was associated with the Wnt3 target gene. CONCLUSION Serum sncRNA analysis shows potential for studying the molecular mechanisms of endometrial receptivity. Four serum tsRNAs can serve as novel biomarkers for non-invasive endometrial receptivity detection. TsRNA-35:73-Asp-GTC-1 may further regulate endometrial receptivity by targeting Wnt3.
Collapse
Affiliation(s)
- Shaoyuan Xu
- Reproductive Medicine Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China; Reproductive Medicine Center, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China; Hubei Clinical Research Center for Reproductive Medicine, Shiyan, Hubei 442000, China
| | - Dongling Hu
- Reproductive Medicine Center, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China; Hubei Clinical Research Center for Reproductive Medicine, Shiyan, Hubei 442000, China
| | - Yanqin Ye
- Reproductive Medicine Center, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China; Hubei Clinical Research Center for Reproductive Medicine, Shiyan, Hubei 442000, China
| | - Yanli Mu
- Reproductive Medicine Center, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China; Hubei Clinical Research Center for Reproductive Medicine, Shiyan, Hubei 442000, China
| | - Yao Xiong
- Reproductive Medicine Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China; Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, Wuhan, Hubei 430071, China.
| | - Yuanzhen Zhang
- Reproductive Medicine Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China; Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, Wuhan, Hubei 430071, China.
| |
Collapse
|
7
|
Kawata Y, Terakawa J, Takeshita A, Namiki T, Kageyama A, Noguchi M, Murakami H, Fukada T, Ito J, Kashiwazaki N. Endometrial zinc transporter Slc39a10/Zip10 is indispensable for progesterone responsiveness and successful pregnancy in mice. PNAS NEXUS 2025; 4:pgaf047. [PMID: 39967682 PMCID: PMC11833700 DOI: 10.1093/pnasnexus/pgaf047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 01/21/2025] [Indexed: 02/20/2025]
Abstract
Zinc is a critical trace element that is important for various biological functions including male and female reproductive systems, but the molecular mechanisms that underlie fertility have been unclear. We show here that zinc signaling in the endometrial tissue is indispensable for successful pregnancy in mice. We observed that a uterine-specific genetic deletion of Slc39a10/Zip10, which encodes one of the zinc transporters to elevate the cytoplasmic level of zinc, results in female infertility due to failure of embryo invasion into the endometrium and subsequent embryonic loss. Zip10 mRNA is expressed in uterine tissues, especially in the decidualizing stromal cells during embryo implantation. The absence of ZIP10 leads to attenuation of progesterone-progesterone receptor (PGR) signals between the epithelium and the stroma, including abnormal expression of the PGR and its target molecules in both the epithelium and stroma in vivo. We found that depletion of intracytoplasmic zinc ions due to loss of ZIP10 disrupts the change in nuclear-to-cytoplasmic localization of GLI1, which is critical for PGR signaling in the decidualizing stromal cells in vitro not only in mice but also in humans. Our findings (i) highlight a biological relevance of ZIP10-mediated zinc homeostatic regulation in the establishment of a successful pregnancy and (ii) will help to prevent infertility in humans.
Collapse
Affiliation(s)
- Yui Kawata
- Laboratory of Animal Reproduction, School of Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara 252-5201, Japan
- Graduate School of Veterinary Science, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara 252-5201, Japan
| | - Jumpei Terakawa
- Graduate School of Veterinary Science, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara 252-5201, Japan
- Laboratory of Toxicology, School of Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara 252-5201, Japan
| | - Ayuu Takeshita
- Laboratory of Animal Reproduction, School of Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara 252-5201, Japan
| | - Takafumi Namiki
- Laboratory of Animal Reproduction, School of Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara 252-5201, Japan
- Graduate School of Veterinary Science, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara 252-5201, Japan
| | - Atsuko Kageyama
- Laboratory of Animal Reproduction, School of Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara 252-5201, Japan
| | - Michiko Noguchi
- Graduate School of Veterinary Science, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara 252-5201, Japan
- Laboratory of Theriogenology, School of Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara 252-5201, Japan
| | - Hironobu Murakami
- Graduate School of Veterinary Science, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara 252-5201, Japan
- Laboratory of Infectious Diseases, School of Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara 252-5201, Japan
| | - Toshiyuki Fukada
- Laboratory of Molecular and Cellular Physiology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihamahouji, Yamashirocho, Tokushima-City, Tokushima 770-8514, Japan
| | - Junya Ito
- Laboratory of Animal Reproduction, School of Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara 252-5201, Japan
- Graduate School of Veterinary Science, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara 252-5201, Japan
| | - Naomi Kashiwazaki
- Laboratory of Animal Reproduction, School of Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara 252-5201, Japan
- Graduate School of Veterinary Science, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara 252-5201, Japan
| |
Collapse
|
8
|
Lee JG, Lee SM, Hyun M, Heo JD. Tire rubber-derived contaminants 6PPD and 6PPD-quinone reduce attachment and outgrowth of trophoblast spheroids onto endometrial epithelial cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117744. [PMID: 39818141 DOI: 10.1016/j.ecoenv.2025.117744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/18/2025]
Abstract
N-(1,3-Dimethylbutyl)-N'-phenyl-p-phenylenediamine (6PPD), a synthetic additive widely used in the rubber industry, and its oxidized product 6PPD-quinone (6PPDQ), have garnered widespread attention as an emerging hazardous chemicals owing to their potential detrimental effects on aquatic ecosystem and human health. The effects of 6PPD and 6PPDq on the female reproductive tract, especially embryo implantation, remain unknown and were investigated in this study. We used the spheroid attachment and outgrowth models of BeWo trophoblastic spheroids and Ishikawa cells as surrogates for the human blastocyst and endometrial epithelium, respectively. Treatment with the chemicals for up to 48 h decreased the viability of the cells in a dose- and cell line-dependent manner (20-100 μM 6PPD and 10-100 μM 6PPDQ for both the cell lines). At a noncytotoxic concentration, exposure of Ishikawa cells to 1 and 10 μM 6PPD reduced the attachment of BeWo spheroids and further inhibited their invasion and outgrowth on the endometrial epithelial monolayer. A similar result was observed in 1 μM 6PPDQ-exposed groups. Gene expression profiling of 6PPD- and 6PPDQ-exposed endometrial epithelial cells revealed that both 6PPD and 6PPDQ differentially regulated a panel of transcript markers toward overall downregulation of receptivity and invasion. The study provides the first proof of the adverse effects of 6PPD and 6PPDQ on human endometrial receptivity and trophoblast invasion during the window of implantation, warranting the need for further in vivo and clinical studies.
Collapse
Affiliation(s)
- Jong Geol Lee
- Center for Bio-Health Research, Division of Gyeongnam Bio-Environmental Research, Korea Institute of Toxicology, Jinju 52834, Republic of Korea.
| | - Seon Min Lee
- Center for Bio-Health Research, Division of Gyeongnam Bio-Environmental Research, Korea Institute of Toxicology, Jinju 52834, Republic of Korea
| | - Moonjung Hyun
- Center for Bio-Health Research, Division of Gyeongnam Bio-Environmental Research, Korea Institute of Toxicology, Jinju 52834, Republic of Korea
| | - Jeong Doo Heo
- Korea Institute of Toxicology, Daejeon 34114, Republic of Korea.
| |
Collapse
|
9
|
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
|
10
|
Fan X, Wei J, Guo Y, Ma J, Qi M, Huang H, Zheng P, Jiang W, Yao Y. LPS Disrupts Endometrial Receptivity by Inhibiting STAT1 Phosphorylation in Sheep. Int J Mol Sci 2024; 25:13673. [PMID: 39769435 PMCID: PMC11678167 DOI: 10.3390/ijms252413673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/05/2024] [Accepted: 12/19/2024] [Indexed: 01/30/2025] Open
Abstract
Uterine infections reduce ruminant reproductive efficiency. Reproductive dysfunction caused by infusion of Gram-negative bacteria is characterized by the failure of embryo implantation and reduced conception rates. Lipopolysaccharide (LPS), a major component of the outer membrane of Gram-negative bacteria, is highly abortogenic. In this study, the effects of LPS infusion on the endometrial receptivity of sheep were studied during three critical periods of embryo implantation. The results showed that LPS infusion on d12, d16, and d20 of pregnancy in vivo interfered with the expression of prostaglandins (PGs) and affected the expression of adhesion-related factors (ITGB1/3/5, SPP1), key implantation genes (HOXA10, HOXA11 and LIF), and progestational elongation genes (ISG15, RSAD2 and CXCL10) during embryo implantation. In addition, after LPS infusion on d12, d16, and d20, the phosphorylation level of STAT1 significantly decreased and the protein expression level of IRF9 significantly increased on d12, suggesting that LPS infusion in sheep impairs endometrial receptivity through the JAK2/STAT1 pathway. Sheep endometrial epithelial cells were treated with 17 β-estrogen, progesterone, and/or interferon-tau in vitro to mimic the receptivity of the endometrium during early pregnancy for validation. LPS and the p-STAT1 inhibitor fludarabine were both added to the model, which resulted in reduced p-STAT1 protein expression, significant inhibition of PGE2/PGF2α, and significant suppression of the expression of key embryo implantation genes. Collectively, these results indicate that LPS infusion in sheep on d12, d16, and d20 impairs endometrial receptivity through the JAK2/STAT1 pathway, which is responsible for LPS-associated pregnancy failure.
Collapse
Affiliation(s)
- Xing Fan
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150038, China; (X.F.); (J.W.); (Y.G.); (J.M.); (H.H.); (P.Z.)
| | - Jinzi Wei
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150038, China; (X.F.); (J.W.); (Y.G.); (J.M.); (H.H.); (P.Z.)
| | - Yu Guo
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150038, China; (X.F.); (J.W.); (Y.G.); (J.M.); (H.H.); (P.Z.)
| | - Juan Ma
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150038, China; (X.F.); (J.W.); (Y.G.); (J.M.); (H.H.); (P.Z.)
| | - Meiyu Qi
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China;
| | - He Huang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150038, China; (X.F.); (J.W.); (Y.G.); (J.M.); (H.H.); (P.Z.)
| | - Peng Zheng
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150038, China; (X.F.); (J.W.); (Y.G.); (J.M.); (H.H.); (P.Z.)
| | - Wenjie Jiang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150038, China; (X.F.); (J.W.); (Y.G.); (J.M.); (H.H.); (P.Z.)
| | - Yuchang Yao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150038, China; (X.F.); (J.W.); (Y.G.); (J.M.); (H.H.); (P.Z.)
| |
Collapse
|
11
|
Bourdiec A, Messaoudi S, El Kasmi I, Chow-Shi-Yée M, Kadoch E, Stebenne ME, Tadevosyan A, Kadoch IJ. Development of a New Personalized Molecular Test Based on Endometrial Receptivity and Maternal-Fetal Dialogue: Adhesio. Biochem Genet 2024:10.1007/s10528-024-10950-y. [PMID: 39488671 DOI: 10.1007/s10528-024-10950-y] [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: 07/30/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024]
Abstract
Successful embryo implantation relies on a receptive endometrium and a maternofetal dialogue. Abnormal receptivity is a common cause of implantation failure in assisted reproductive techniques. This study aimed to develop a novel transcriptomic-based diagnostic assay, Adhesio, for assessing endometrial receptivity and guiding personalized embryo transfer. Adhesio was developed based on an initial dataset of 74 endometrial biopsies. Two types of biopsy samples were involved: 45 endometrial biopsies collected during the optimal theoretical window of implantation (WOI) and 29 endometrial biopsies which cells have been cultured with or without an autologous embryo. Microarray analysis was performed to identify differentially expressed genes associated with endometrial receptivity and selected candidate genes were assessed using quantitative real-time polymerase chain reaction (RT-qPCR) on biopsy samples. Statistical analyses were conducted to assess the performance and accuracy of Adhesio. The microarray analysis identified three distinct clusters of endometrial samples with differential gene expression patterns. Cluster 1 exhibited 1717 differentially expressed genes involved in biological processes associated with endometrial receptivity. A specific transcriptomic signature of 60 genes associated with endometrial co-culture was obtained using class prediction approach. Thereafter, an original panel of 10 genes was selected as potential biomarkers for endometrial receptivity based on their expression profiles in both endometrial biopsies and co-cultured cells. This article outlines the methodology employed to develop Adhesio, a test that assesses endometrial receptivity using an original panel of 10 genes. These genes are not only involved during the WOI but are also influenced by the maternal-fetal dialogue.
Collapse
Affiliation(s)
- Amelie Bourdiec
- Clinique ovo, 8000 Boul. Décarie, Montreal, QC, H4P 2S4, Canada
| | | | - Imane El Kasmi
- Clinique ovo, 8000 Boul. Décarie, Montreal, QC, H4P 2S4, Canada
| | | | - Eva Kadoch
- Clinique ovo, 8000 Boul. Décarie, Montreal, QC, H4P 2S4, Canada
| | | | - Artak Tadevosyan
- Clinique ovo, 8000 Boul. Décarie, Montreal, QC, H4P 2S4, Canada
- Department of Pharmacology and Physiology, Université de Montreal, Montreal, QC, Canada
| | - Isaac-Jacques Kadoch
- Clinique ovo, 8000 Boul. Décarie, Montreal, QC, H4P 2S4, Canada.
- Department of Obstetrics and Gynecology, Université de Montreal, Montreal, QC, Canada.
| |
Collapse
|
12
|
Kleinová M, Varga I, Čeháková M, Valent M, Klein M. Exploring the black box of human reproduction: endometrial organoids and assembloids - generation, implantation modeling, and future clinical perspectives. Front Cell Dev Biol 2024; 12:1482054. [PMID: 39507423 PMCID: PMC11539068 DOI: 10.3389/fcell.2024.1482054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024] Open
Abstract
One of the critical processes in human reproduction that is still poorly understood is implantation. The implantation of an early human embryo is considered a significant limitation of successful pregnancy. Therefore, researchers are trying to develop an ideal model of endometrium in vitro that can mimic the endometrial micro-environment in vivo as much as possible. The ultimate goal of endometrial modeling is to study the molecular interactions at the embryo-maternal interface and to use this model as an in vitro diagnostic tool for infertility. Significant progress has been made over the years in generating such models. The first experiments of endometrial modeling involved animal models, which are undoubtedly valuable, but at the same time, their dissimilarities with human tissue represent a significant obstacle to further research. This fact led researchers to develop basic monolayer coculture systems using uterine cells obtained from biopsies and, later on, complex and multilayer coculture models. With successful tissue engineering methods and various cultivation systems, it is possible to form endometrial two-dimensional (2D) models to three-dimensional (3D) organoids and novel assembloids that can recapitulate many aspects of endometrial tissue architecture and cell composition. These organoids have already helped to provide new insight into the embryo-endometrium interplay. The main aim of this paper is a comprehensive review of past and current approaches to endometrial model generation, their feasibility, and potential clinical application for infertility treatment.
Collapse
Affiliation(s)
- Mária Kleinová
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Ivan Varga
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Michaela Čeháková
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| | - Martin Valent
- Department of Gynecology and Obstetrics, University Hospital Bratislava – Kramáre Workplace, Bratislava, Slovakia
| | - Martin Klein
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovakia
| |
Collapse
|
13
|
Jefferson W, Wang T, Padilla-Banks E, Williams C. Unexpected nuclear hormone receptor and chromatin dynamics regulate estrous cycle dependent gene expression. Nucleic Acids Res 2024; 52:10897-10917. [PMID: 39166489 PMCID: PMC11472041 DOI: 10.1093/nar/gkae714] [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: 01/15/2024] [Revised: 06/27/2024] [Accepted: 08/06/2024] [Indexed: 08/23/2024] Open
Abstract
Chromatin changes in response to estrogen and progesterone are well established in cultured cells, but how they control gene expression under physiological conditions is largely unknown. To address this question, we examined in vivo estrous cycle dynamics of mouse uterus hormone receptor occupancy, chromatin accessibility and chromatin structure by combining RNA-seq, ATAC-seq, HiC-seq and ChIP-seq. Two estrous cycle stages were chosen for these analyses, diestrus (highest estrogen) and estrus (highest progesterone). Unexpectedly, rather than alternating with each other, estrogen receptor alpha (ERα) and progesterone receptor (PGR) were co-bound during diestrus and lost during estrus. Motif analysis of open chromatin followed by hypoxia inducible factor 2A (HIF2A) ChIP-seq and conditional uterine deletion of this transcription factor revealed a novel role for HIF2A in regulating diestrus gene expression patterns that were independent of either ERα or PGR binding. Proteins in complex with ERα included PGR and cohesin, only during diestrus. Combined with HiC-seq analyses, we demonstrate that complex chromatin architecture changes including enhancer switching are coordinated with ERα and PGR co-binding during diestrus and non-hormone receptor transcription factors such as HIF2A during estrus to regulate most differential gene expression across the estrous cycle.
Collapse
Affiliation(s)
- Wendy N Jefferson
- Reproductive & Developmental Biology Laboratory, Research Triangle Park, NC 27709, USA
| | - Tianyuan Wang
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA
| | | | - Carmen J Williams
- Reproductive & Developmental Biology Laboratory, Research Triangle Park, NC 27709, USA
| |
Collapse
|
14
|
Akaeda S, Aikawa S, Hirota Y. Spatial and molecular anatomy of the endometrium during embryo implantation: a current overview of key regulators of blastocyst invasion. FEBS J 2024; 291:4206-4221. [PMID: 38348632 DOI: 10.1111/febs.17077] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 01/09/2024] [Accepted: 01/23/2024] [Indexed: 10/04/2024]
Abstract
Embryo implantation is composed of three steps: blastocyst apposition, adhesion/attachment and invasion. Blastocyst invasion has been studied less extensively than the other two events. Historically, studies conducted using electron microscopy have shown the removal of epithelial cells in the vicinity of the attached blastocysts in rodents, although the underlying mechanisms have remained unclear. Here, we describe recent studies using mice with uterine-specific gene deletion that demonstrated important roles for nuclear proteins such as progesterone receptor, hypoxia inducible factor and retinoblastoma in the regulation of embryo invasion. In these mouse models, the detachment of the endometrial luminal epithelium, decidualization in the stroma, and the activation of trophoblasts have been found to be important in ensuring embryo invasion. This review summarizes the molecular signaling associated with these cellular events, mainly evidenced by mouse models.
Collapse
Affiliation(s)
- Shun Akaeda
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Shizu Aikawa
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Japan
| | - Yasushi Hirota
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Japan
| |
Collapse
|
15
|
Santini L, Kowald S, Cerron-Alvan LM, Huth M, Fabing AP, Sestini G, Rivron N, Leeb M. FoxO transcription factors actuate the formative pluripotency specific gene expression programme. Nat Commun 2024; 15:7879. [PMID: 39251582 PMCID: PMC11384738 DOI: 10.1038/s41467-024-51794-9] [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: 01/24/2024] [Accepted: 08/16/2024] [Indexed: 09/11/2024] Open
Abstract
Naïve pluripotency is sustained by a self-reinforcing gene regulatory network (GRN) comprising core and naïve pluripotency-specific transcription factors (TFs). Upon exiting naïve pluripotency, embryonic stem cells (ESCs) transition through a formative post-implantation-like pluripotent state, where they acquire competence for lineage choice. However, the mechanisms underlying disengagement from the naïve GRN and initiation of the formative GRN are unclear. Here, we demonstrate that phosphorylated AKT acts as a gatekeeper that prevents nuclear localisation of FoxO TFs in naïve ESCs. PTEN-mediated reduction of AKT activity upon exit from naïve pluripotency allows nuclear entry of FoxO TFs, enforcing a cell fate transition by binding and activating formative pluripotency-specific enhancers. Indeed, FoxO TFs are necessary and sufficient for the activation of the formative pluripotency-specific GRN. Our work uncovers a pivotal role for FoxO TFs in establishing formative post-implantation pluripotency, a critical early embryonic cell fate transition.
Collapse
Affiliation(s)
- Laura Santini
- Max Perutz Laboratories Vienna, University of Vienna, Vienna BioCenter, 1030, Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna, Medical University of Vienna, 1030, Vienna, Austria
| | - Saskia Kowald
- Max Perutz Laboratories Vienna, University of Vienna, Vienna BioCenter, 1030, Vienna, Austria
| | - Luis Miguel Cerron-Alvan
- Max Perutz Laboratories Vienna, University of Vienna, Vienna BioCenter, 1030, Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna, Medical University of Vienna, 1030, Vienna, Austria
| | - Michelle Huth
- Max Perutz Laboratories Vienna, University of Vienna, Vienna BioCenter, 1030, Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna, Medical University of Vienna, 1030, Vienna, Austria
| | - Anna Philina Fabing
- Max Perutz Laboratories Vienna, University of Vienna, Vienna BioCenter, 1030, Vienna, Austria
| | - Giovanni Sestini
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna, Medical University of Vienna, 1030, Vienna, Austria
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, 1030, Vienna, Austria
| | - Nicolas Rivron
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, 1030, Vienna, Austria
| | - Martin Leeb
- Max Perutz Laboratories Vienna, University of Vienna, Vienna BioCenter, 1030, Vienna, Austria.
| |
Collapse
|
16
|
Li MY, Shen HH, Cao XY, Gao XX, Xu FY, Ha SY, Sun JS, Liu SP, Xie F, Li MQ. Targeting a mTOR/autophagy axis: a double-edged sword of rapamycin in spontaneous miscarriage. Biomed Pharmacother 2024; 177:116976. [PMID: 38906022 DOI: 10.1016/j.biopha.2024.116976] [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: 03/29/2024] [Revised: 06/02/2024] [Accepted: 06/15/2024] [Indexed: 06/23/2024] Open
Abstract
Immune dysfunction is a primary culprit behind spontaneous miscarriage (SM). To address this, immunosuppressive agents have emerged as a novel class of tocolytic drugs, modulating the maternal immune system's tolerance towards the embryo. Rapamycin (PubChem CID:5284616), a dual-purpose compound, functions as an immunosuppressive agent and triggers autophagy by targeting the mTOR pathway. Its efficacy in treating SM has garnered significant research interest in recent times. Autophagy, the cellular process of self-degradation and recycling, plays a pivotal role in numerous health conditions. Research indicates that autophagy is integral to endometrial decidualization, trophoblast invasion, and the proper functioning of decidual immune cells during a healthy pregnancy. Yet, in cases of SM, there is a dysregulation of the mTOR/autophagy axis in decidual stromal cells or immune cells at the maternal-fetal interface. Both in vitro and in vivo studies have highlighted the potential benefits of low-dose rapamycin in managing SM. However, given mTOR's critical role in energy metabolism, inhibiting it could potentially harm the pregnancy. Moreover, while low-dose rapamycin has been deemed safe for treating recurrent implant failure, its potential teratogenic effects remain uncertain due to insufficient data. In summary, rapamycin represents a double-edged sword in the treatment of SM, balancing its impact on autophagy and immune regulation. Further investigation is warranted to fully understand its implications.
Collapse
Affiliation(s)
- Meng-Ying Li
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
| | - Hui-Hui Shen
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
| | - Xiao-Yan Cao
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
| | - Xiao-Xiao Gao
- Department of Obstetrics and Gynecology, Jinshan Hospital, Fudan University, Shanghai 201508, People's Republic of China
| | - Feng-Yuan Xu
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States
| | - Si-Yao Ha
- Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510235, China
| | - Jian-Song Sun
- School of Life Science and Health Engineering, Jiangnan University, Wuxi 214122, People's Republic of China
| | - Song-Ping Liu
- Department of Obstetrics and Gynecology, Jinshan Hospital, Fudan University, Shanghai 201508, People's Republic of China.
| | - Feng Xie
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People's Republic of China.
| | - Ming-Qing Li
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China; Department of Gynecologic Endocrinology and Reproductive Immunology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai 200030, People's Republic of China.
| |
Collapse
|
17
|
Diaz-Gimeno P, Sebastian-Leon P, Spath K, Marti-Garcia D, Sanchez-Reyes JM, Vidal MDC, Devesa-Peiro A, Sanchez-Ribas I, Martinez-Martinez A, Pellicer N, Wells D, Pellicer A. Predicting risk of endometrial failure: a biomarker signature that identifies a novel disruption independent of endometrial timing in patients undergoing hormonal replacement cycles. Fertil Steril 2024; 122:352-364. [PMID: 38518993 DOI: 10.1016/j.fertnstert.2024.03.015] [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: 05/17/2023] [Revised: 03/14/2024] [Accepted: 03/18/2024] [Indexed: 03/24/2024]
Abstract
OBJECTIVE To propose a new gene expression signature that identifies endometrial disruptions independent of endometrial luteal phase timing and predicts if patients are at risk of endometrial failure. DESIGN Multicentric, prospective study. SETTING Reproductive medicine research department in a public hospital affiliated with private fertility clinics and a reproductive genetics laboratory. PATIENTS Caucasian women (n = 281; 39.4 ± 4.8 years old with a body mass index of 22.9 ± 3.5 kg/m2) undergoing hormone replacement therapy between July 2018 and July 2021. Endometrial samples from 217 patients met RNA quality criteria for signature discovery and analysis. INTERVENTION(S) Endometrial biopsies collected in the mid-secretory phase. MAIN OUTCOME MEASURE(S) Endometrial luteal phase timing-corrected expression of 404 genes and reproductive outcomes of the first single embryo transfer (SET) after biopsy collection to identify prognostic biomarkers of endometrial failure. RESULTS Removal of endometrial timing variation from gene expression data allowed patients to be stratified into poor (n = 137) or good (n = 49) endometrial prognosis groups on the basis of their clinical and transcriptomic profiles. Significant differences were found between endometrial prognosis groups in terms of reproductive rates: pregnancy (44.6% vs. 79.6%), live birth (25.6% vs. 77.6%), clinical miscarriage (22.2% vs. 2.6%), and biochemical miscarriage (20.4% vs. 0%). The relative risk of endometrial failure for patients predicted as a poor endometrial prognosis was 3.3 times higher than those with a good prognosis. The differences in gene expression between both profiles were proposed as a biomarker, coined the endometrial failure risk (EFR) signature. Poor prognosis profiles were characterized by 59 upregulated and 63 downregulated genes mainly involved in regulation (17.0%), metabolism (8.4%), immune response, and inflammation (7.8%). This EFR signature had a median accuracy of 0.92 (min = 0.88, max = 0.94), median sensitivity of 0.96 (min = 0.91, max = 0.98), and median specificity of 0.84 (min = 0.77, max = 0.88), positioning itself as a promising biomarker for endometrial evaluation. CONCLUSION(S) The EFR signature revealed a novel endometrial disruption, independent of endometrial luteal phase timing, present in 73.7% of patients. This EFR signature stratified patients into 2 significantly distinct and clinically relevant prognosis profiles providing opportunities for personalized therapy. Nevertheless, further validations are needed before implementing this gene signature as an artificial intelligence (AI)-based tool to reduce the risk of patients experiencing endometrial failure.
Collapse
Affiliation(s)
- Patricia Diaz-Gimeno
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain.
| | - Patricia Sebastian-Leon
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | | | - Diana Marti-Garcia
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Josefa Maria Sanchez-Reyes
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain; Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, Valencia, Spain
| | - Maria Del Carmen Vidal
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain; Reproductive Medicine Center, IVI RMA Valencia, Valencia, Spain
| | - Almudena Devesa-Peiro
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain; Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, Valencia, Spain
| | - Immaculada Sanchez-Ribas
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain; Reproductive Medicine Center, IVI RMA Barcelona, Barcelona, Spain
| | - Asunta Martinez-Martinez
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Nuria Pellicer
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain; Reproductive Medicine Center, IVI RMA Valencia, Valencia, Spain
| | - Dagan Wells
- JUNO Genetics, Winchester House, Oxford, United Kingdom; Nuffield Department of Women's & Reproductive Health, University of Oxford, Women's Centre John Radcliffe Hospital, Oxford, United Kingdom
| | - Antonio Pellicer
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain; JUNO Genetics, Winchester House, Oxford, United Kingdom; Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, Valencia, Spain; Reproductive Medicine Center, IVI RMA Rome, Largo Il de brando Pizzetti, Roma, Italy
| |
Collapse
|
18
|
Deng N, Li G, Zhang L, Wang P, Liu M, He B, Tang Y, Cai H, Lu J, Wang H, Deng W, Bao H, Kong S. H3K27me3 timely dictates uterine epithelial transcriptome remodeling and thus transformation essential for normal embryo implantation. Cell Death Differ 2024; 31:1013-1028. [PMID: 38698061 PMCID: PMC11303564 DOI: 10.1038/s41418-024-01302-9] [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: 12/11/2023] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 05/05/2024] Open
Abstract
Uterine luminal epithelia (LE), the first layer contacting with the blastocyst, acquire receptivity for normal embryo implantation. Besides the well-accepted transcriptional regulation dominated by ovarian estrogen and progesterone for receptivity establishment, the involvement of epigenetic mechanisms remains elusive. This study systematically profiles the transcriptome and genome-wide H3K27me3 distribution in the LE throughout the preimplantation. Combining genetic and pharmacological approaches targeting the PRC2 core enzyme Ezh1/2, we demonstrate that the defective remodeling of H3K27me3 in the preimplantation stage disrupts the differentiation of LE, and derails uterine receptivity, resulting in implantation failure. Specifically, crucial epithelial genes, Pgr, Gata2, and Sgk1, are transcriptionally silenced through de novo deposition of H3K27me3 for LE transformation, and their sustained expression in the absence of H3K27me3 synergistically confines the nuclear translocation of FOXO1. Further functional studies identify several actin-associated genes, including Arpin, Tmod1, and Pdlim2, as novel direct targets of H3K27me3. Their aberrantly elevated expression impedes the morphological remodeling of LE, a hindrance alleviated by treatment with cytochalasin D which depolymerizes F-actin. Collectively, this study uncovers a previously unappreciated epigenetic regulatory mechanism for the transcriptional silencing of key LE genes via H3K27me3, essential for LE differentiation and thus embryo implantation.
Collapse
Affiliation(s)
- Na Deng
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Gaizhen Li
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Leilei Zhang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Peiran Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Mengying Liu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Bo He
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Yedong Tang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Han Cai
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Jinhua Lu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Haibin Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China.
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China.
| | - Wenbo Deng
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China.
| | - Haili Bao
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China.
| | - Shuangbo Kong
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China.
| |
Collapse
|
19
|
Hong L, Xiao S, Diao L, Lian R, Chen C, Zeng Y, Liu S. Decreased AMPK/SIRT1/PDK4 induced by androgen excess inhibits human endometrial stromal cell decidualization in PCOS. Cell Mol Life Sci 2024; 81:324. [PMID: 39080028 PMCID: PMC11335245 DOI: 10.1007/s00018-024-05362-5] [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: 01/28/2024] [Revised: 07/04/2024] [Accepted: 07/11/2024] [Indexed: 08/22/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a complex common endocrine disorder affecting women of reproductive age. Ovulatory dysfunction is recognized as a primary infertile factor, however, even when ovulation is medically induced and restored, PCOS patients continue to experience reduced cumulative pregnancy rates and a higher spontaneous miscarriage rate. Hyperandrogenism, a hallmark feature of PCOS, affects ovarian folliculogenesis, endometrial receptivity, and the establishment and maintenance of pregnancy. Decidualization denotes the transformation that the stromal compart of the endometrium must undergo to accommodate pregnancy, driven by the rising progesterone levels and local cAMP production. However, studies on the impact of hyperandrogenism on decidualization are limited. In this study, we observed that primary endometrial stromal cells from women with PCOS exhibit abnormal responses to progesterone during in vitro decidualization. A high concentration of testosterone inhibits human endometrial stromal cells (HESCs) decidualization. RNA-Seq analysis demonstrated that pyruvate dehydrogenase kinase 4 (PDK4) expression was significantly lower in the endometrium of PCOS patients with hyperandrogenism compared to those without hyperandrogenism. We also characterized that the expression of PDK4 is elevated in the endometrium stroma at the mid-secretory phase. Artificial decidualization could enhance PDK4 expression, while downregulation of PDK4 leads to abnormal decidualization both in vivo and in vitro. Mechanistically, testosterone excess inhibits IGFBP1 and PRL expression, followed by phosphorylating of AMPK that stimulates PDK4 expression. Based on co-immunoprecipitation analysis, we observed an interaction between SIRT1 and PDK4, promoting glycolysis to facilitate decidualization. Restrain of AR activation resumes the AMPK/SIRT1/PDK4 pathway suppressed by testosterone excess, indicating that testosterone primarily acts on decidualization through AR stimulation. Androgen excess in the endometrium inhibits decidualization by disrupting the AMPK/SIRT1/PDK4 signaling pathway. These data demonstrate the critical roles of endometrial PDK4 in regulating decidualization and provide valuable information for understanding the underlying mechanism during decidualization.
Collapse
Affiliation(s)
- Ling Hong
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (Formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Guangdong, China
| | - Shan Xiao
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (Formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, China
| | - Lianghui Diao
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (Formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Guangdong, China
| | - Ruochun Lian
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (Formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, China
| | - Cong Chen
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (Formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, China
| | - Yong Zeng
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (Formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Guangdong, China
| | - Su Liu
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics and Gynecology Hospital (Formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, China.
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Guangdong, China.
| |
Collapse
|
20
|
Dias Da Silva I, Wuidar V, Zielonka M, Pequeux C. Unraveling the Dynamics of Estrogen and Progesterone Signaling in the Endometrium: An Overview. Cells 2024; 13:1236. [PMID: 39120268 PMCID: PMC11312103 DOI: 10.3390/cells13151236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 06/25/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024] Open
Abstract
The endometrium is crucial for the perpetuation of human species. It is a complex and dynamic tissue lining the inner wall of the uterus, regulated throughout a woman's life based on estrogen and progesterone fluctuations. During each menstrual cycle, this multicellular tissue undergoes cyclical changes, including regeneration, differentiation in order to allow egg implantation and embryo development, or shedding of the functional layer in the absence of pregnancy. The biology of the endometrium relies on paracrine interactions between epithelial and stromal cells involving complex signaling pathways that are modulated by the variations of estrogen and progesterone levels across the menstrual cycle. Understanding the complexity of estrogen and progesterone receptor signaling will help elucidate the mechanisms underlying normal reproductive physiology and provide fundamental knowledge contributing to a better understanding of the consequences of hormonal imbalances on gynecological conditions and tumorigenesis. In this narrative review, we delve into the physiology of the endometrium, encompassing the complex signaling pathways of estrogen and progesterone.
Collapse
Grants
- J.0165.24, 7.6529.23, J.0153.22, 7.4580.21F, 7.6518.21, J.0131.19 Fund for Scientific Research
- FSR-F-2023-FM, FSR-F-2022-FM, FSR-F-2021-FM, FSR-F-M-19/6761 University of Liège
- 2020, 2021, 2022 Fondation Léon Fredericq
Collapse
Affiliation(s)
| | | | | | - Christel Pequeux
- Tumors and Development, Estrogen-Sensitive Tissues and Cancer Team, GIGA-Cancer, Laboratory of Biology, University of Liège, 4000 Liège, Belgium; (I.D.D.S.); (V.W.); (M.Z.)
| |
Collapse
|
21
|
Wang B, Gao M, Yao Y, Shen H, Li H, Sun J, Wang L, Zhang X. Enhancing endometrial receptivity: the roles of human chorionic gonadotropin in autophagy and apoptosis regulation in endometrial stromal cells. Reprod Biol Endocrinol 2024; 22:37. [PMID: 38576003 PMCID: PMC10993617 DOI: 10.1186/s12958-024-01205-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 03/12/2024] [Indexed: 04/06/2024] Open
Abstract
Inadequate endometrial receptivity often results in embryo implantation failure and miscarriage. Human chorionic gonadotropin (hCG) is a key signaling molecule secreted during early embryonic development, which regulates embryonic maternal interface signaling and promotes embryo implantation. This study aimed to examine the impact of hCG on endometrial receptivity and its underlying mechanisms. An exploratory study was designed, and endometrial samples were obtained from women diagnosed with simple tubal infertility or male factor infertile (n = 12) and recurrent implantation failure (RIF, n = 10). Using reverse transcription-quantitative PCR and western blotting, luteinizing hormone (LH)/hCG receptor (LHCGR) levels and autophagy were detected in the endometrial tissues. Subsequently, primary endometrial stromal cells (ESCs) were isolated from these control groups and treated with hCG to examine the presence of LHCGR and markers of endometrial receptivity (HOXA10, ITGB3, FOXO1, LIF, and L-selectin ligand) and autophagy-related factors (Beclin1, LC3, and P62). The findings revealed that the expressions of receptivity factors, LHCGR, and LC3 were reduced in the endometrial tissues of women with RIF compared with the control group, whereas the expression of P62 was elevated. The administration of hCG to ESCs specifically activated LHCGR, stimulating an increase in the endometrial production of HOXA10, ITGB3, FOXO1, LIF and L-selectin ligands. Furthermore, when ESCs were exposed to 0.1 IU/mL hCG for 72 h, the autophagy factors Beclin1 and LC3 increased within the cells and P62 decreased. Moreover, the apoptotic factor Bax increased and Bcl-2 declined. However, when small interfering RNA was used to knock down LHCGR, hCG was less capable of controlling endometrial receptivity and autophagy molecules in ESCs. In addition, hCG stimulation enhanced the phosphorylation of ERK1/2 and mTOR proteins. These results suggest that women with RIF exhibit lower levels of LHCGR and compromised autophagy function in their endometrial tissues. Thus, hCG/LHCGR could potentially improve endometrial receptivity by modulating autophagy and apoptosis.
Collapse
Affiliation(s)
- Bin Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Mingxia Gao
- Reproductive Medicine Center, The First Hospital of Lanzhou University, Lanzhou, China.
- Key Laboratory for Reproductive Medicine and Embryo, Gansu Province, Lanzhou, China.
| | - Ying Yao
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Haofei Shen
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Hongwei Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Jingjing Sun
- Medical Laboratory Center, The First Hospital of Lanzhou University, Lanzhou, China
| | - Liyan Wang
- Reproductive Medicine Center, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory for Reproductive Medicine and Embryo, Gansu Province, Lanzhou, China
| | - Xuehong Zhang
- Reproductive Medicine Center, The First Hospital of Lanzhou University, Lanzhou, China.
- Key Laboratory for Reproductive Medicine and Embryo, Gansu Province, Lanzhou, China.
| |
Collapse
|
22
|
Liao Z, Tang S, Nozawa K, Shimada K, Ikawa M, Monsivais D, Matzuk M. Affinity-tagged SMAD1 and SMAD5 mouse lines reveal transcriptional reprogramming mechanisms during early pregnancy. eLife 2024; 12:RP91434. [PMID: 38536963 PMCID: PMC10972565 DOI: 10.7554/elife.91434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024] Open
Abstract
Endometrial decidualization, a prerequisite for successful pregnancies, relies on transcriptional reprogramming driven by progesterone receptor (PR) and bone morphogenetic protein (BMP)-SMAD1/SMAD5 signaling pathways. Despite their critical roles in early pregnancy, how these pathways intersect in reprogramming the endometrium into a receptive state remains unclear. To define how SMAD1 and/or SMAD5 integrate BMP signaling in the uterus during early pregnancy, we generated two novel transgenic mouse lines with affinity tags inserted into the endogenous SMAD1 and SMAD5 loci (Smad1HA/HA and Smad5PA/PA). By profiling the genome-wide distribution of SMAD1, SMAD5, and PR in the mouse uterus, we demonstrated the unique and shared roles of SMAD1 and SMAD5 during the window of implantation. We also showed the presence of a conserved SMAD1, SMAD5, and PR genomic binding signature in the uterus during early pregnancy. To functionally characterize the translational aspects of our findings, we demonstrated that SMAD1/5 knockdown in human endometrial stromal cells suppressed expressions of canonical decidual markers (IGFBP1, PRL, FOXO1) and PR-responsive genes (RORB, KLF15). Here, our studies provide novel tools to study BMP signaling pathways and highlight the fundamental roles of SMAD1/5 in mediating both BMP signaling pathways and the transcriptional response to progesterone (P4) during early pregnancy.
Collapse
Affiliation(s)
- Zian Liao
- Department of Pathology & Immunology, Baylor College of MedicineHoustonUnited States
- Graduate Program of Genetics and Genomics, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
- Center for Drug Discovery, Baylor College of MedicineHoustonUnited States
| | - Suni Tang
- Department of Pathology & Immunology, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Kaori Nozawa
- Department of Pathology & Immunology, Baylor College of MedicineHoustonUnited States
- Center for Drug Discovery, Baylor College of MedicineHoustonUnited States
| | - Keisuke Shimada
- Research Institute for Microbial Diseases, Osaka UniversityOsakaJapan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka UniversityOsakaJapan
| | - Diana Monsivais
- Department of Pathology & Immunology, Baylor College of MedicineHoustonUnited States
- Center for Drug Discovery, Baylor College of MedicineHoustonUnited States
| | - Martin Matzuk
- Department of Pathology & Immunology, Baylor College of MedicineHoustonUnited States
- Graduate Program of Genetics and Genomics, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
- Center for Drug Discovery, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
23
|
Yan J, Zhou L, Liu M, Zhu H, Zhang X, Cai E, Xu X, Chen T, Cheng H, Liu J, Wang S, Dai L, Chang X, Tang F. Single-cell analysis reveals insights into epithelial abnormalities in ovarian endometriosis. Cell Rep 2024; 43:113716. [PMID: 38412094 DOI: 10.1016/j.celrep.2024.113716] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/19/2023] [Accepted: 01/11/2024] [Indexed: 02/29/2024] Open
Abstract
Ovarian endometriosis is characterized by the growth of endometrial tissue within the ovary, causing infertility and chronic pain. However, its pathophysiology remains unclear. Utilizing high-precision single-cell RNA sequencing, we profile the normal, eutopic, and ectopic endometrium from 34 individuals across proliferative and secretory phases. We observe an increased proportion of ciliated cells in both eutopic and ectopic endometrium, characterized by a diminished expression of estrogen sulfotransferase, which likely confers apoptosis resistance. After translocating to ectopic lesions, endometrial epithelium upregulates nicotinamide N-methyltransferase expression that inhibits apoptosis by promoting deacetylation and subsequent nuclear exclusion of transcription factor forkhead box protein O1, thereby leading to the downregulation of the apoptotic gene BIM. Moreover, epithelial cells in ectopic lesions elevate HLA class II complex expression, which stimulates CD4+ T cells and consequently contributes to chronic inflammation. Altogether, our study provides a comprehensive atlas of ovarian endometriosis and highlights potential therapeutic targets for modulating apoptosis and inflammation.
Collapse
Affiliation(s)
- Jia Yan
- School of Life Sciences, Biomedical Pioneering Innovation Center, Peking University, Beijing 100000, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100000, China; Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Peking University, Beijing 100000, China
| | - Ling Zhou
- Department of Obstetrics and Gynecology, People's Hospital, Peking University, Beijing 100044, China
| | - Mengya Liu
- School of Life Sciences, Biomedical Pioneering Innovation Center, Peking University, Beijing 100000, China; Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Peking University, Beijing 100000, China
| | - Honglan Zhu
- Department of Obstetrics and Gynecology, People's Hospital, Peking University, Beijing 100044, China
| | - Xin Zhang
- School of Life Sciences, Biomedical Pioneering Innovation Center, Peking University, Beijing 100000, China
| | - E Cai
- Department of Obstetrics and Gynecology, People's Hospital, Peking University, Beijing 100044, China
| | - Xueqiang Xu
- School of Life Sciences, Biomedical Pioneering Innovation Center, Peking University, Beijing 100000, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100000, China; Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Peking University, Beijing 100000, China
| | - Tinghan Chen
- School of Life Sciences, Biomedical Pioneering Innovation Center, Peking University, Beijing 100000, China
| | - Hongyan Cheng
- Department of Obstetrics and Gynecology, People's Hospital, Peking University, Beijing 100044, China
| | - Jun'e Liu
- School of Life Sciences, Biomedical Pioneering Innovation Center, Peking University, Beijing 100000, China; Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Peking University, Beijing 100000, China
| | - Shang Wang
- Department of Obstetrics and Gynecology, People's Hospital, Peking University, Beijing 100044, China
| | - Lin Dai
- Department of Pathology, People's Hospital, Peking University, Beijing 100044, China
| | - Xiaohong Chang
- Department of Obstetrics and Gynecology, People's Hospital, Peking University, Beijing 100044, China.
| | - Fuchou Tang
- School of Life Sciences, Biomedical Pioneering Innovation Center, Peking University, Beijing 100000, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100000, China; Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Peking University, Beijing 100000, China.
| |
Collapse
|
24
|
Garg A, Zielinska AP, Yeung AC, Abdelmalak R, Chen R, Hossain A, Israni A, Nelson SM, Babwah AV, Dhillo WS, Abbara A. Luteal phase support in assisted reproductive technology. Nat Rev Endocrinol 2024; 20:149-167. [PMID: 38110672 DOI: 10.1038/s41574-023-00921-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/31/2023] [Indexed: 12/20/2023]
Abstract
Infertility affects one in six couples, with in vitro fertilization (IVF) offering many the chance of conception. Compared to the solitary oocyte produced during the natural menstrual cycle, the supraphysiological ovarian stimulation needed to produce multiple oocytes during IVF results in a dysfunctional luteal phase that can be insufficient to support implantation and maintain pregnancy. Consequently, hormonal supplementation with luteal phase support, principally exogenous progesterone, is used to optimize pregnancy rates; however, luteal phase support remains largely 'black-box' with insufficient clarity regarding the optimal timing, dosing, route and duration of treatment. Herein, we review the evidence on luteal phase support and highlight remaining uncertainties and future research directions. Specifically, we outline the physiological luteal phase, which is regulated by progesterone from the corpus luteum, and evaluate how it is altered by the supraphysiological ovarian stimulation used during IVF. Additionally, we describe the effects of the hormonal triggers used to mature oocytes on the degree of luteal phase support required. We explain the histological transformation of the endometrium during the luteal phase and evaluate markers of endometrial receptivity that attempt to identify the 'window of implantation'. We also cover progesterone receptor signalling, circulating progesterone levels associated with implantation, and the pharmacokinetics of available progesterone formulations to inform the design of luteal phase support regimens.
Collapse
Affiliation(s)
- Akanksha Garg
- Section of Endocrinology and Investigative Medicine, Imperial College London, Hammersmith Hospital, London, UK
- Imperial College Healthcare NHS Trust, London, UK
| | - Agata P Zielinska
- Section of Endocrinology and Investigative Medicine, Imperial College London, Hammersmith Hospital, London, UK
- Imperial College Healthcare NHS Trust, London, UK
| | - Arthur C Yeung
- Section of Endocrinology and Investigative Medicine, Imperial College London, Hammersmith Hospital, London, UK
- Imperial College Healthcare NHS Trust, London, UK
| | - Rebecca Abdelmalak
- Section of Endocrinology and Investigative Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | - Runzhi Chen
- Section of Endocrinology and Investigative Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | - Aleena Hossain
- Section of Endocrinology and Investigative Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | - Alisha Israni
- Section of Endocrinology and Investigative Medicine, Imperial College London, Hammersmith Hospital, London, UK
| | - Scott M Nelson
- School of Medicine, Dentistry & Nursing, University of Glasgow, Glasgow, UK
- NIHR Biomedical Research Centre, University of Bristol, Bristol, UK
- The Fertility Partnership (TFP), Oxford, UK
| | - Andy V Babwah
- Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Waljit S Dhillo
- Section of Endocrinology and Investigative Medicine, Imperial College London, Hammersmith Hospital, London, UK
- Imperial College Healthcare NHS Trust, London, UK
| | - Ali Abbara
- Section of Endocrinology and Investigative Medicine, Imperial College London, Hammersmith Hospital, London, UK.
- Imperial College Healthcare NHS Trust, London, UK.
| |
Collapse
|
25
|
Zhong Y, Liu F, Zhang X, Guo Q, Wang Z, Wang R. Research progress on reproductive system damage caused by high altitude hypoxia. Endocrine 2024; 83:559-570. [PMID: 38170433 DOI: 10.1007/s12020-023-03643-w] [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: 01/05/2023] [Accepted: 11/28/2023] [Indexed: 01/05/2024]
Abstract
PURPOSE The high altitude area is characterized by low pressure and hypoxia, and rapidly entering the high altitude area will cause a series of damage to the body. Some studies have shown that hypoxia can cause damage to the reproductive system. In recent years, researchers have been paying attention to the effects of hypoxia on hormone level, ovarian reserve, embryonic development, testicular development, sperm motility level, and have begun to explore its injury mechanism, but its mechanism is not clear. In this paper, the mechanism of hypoxia on the reproductive system is reviewed, which is expected to provide a new idea for solving the problem of the low fertility rate of humans and animals at high altitudes. METHODS A comprehensive PubMed search was conducted, selecting all relevant peer-reviewed English papers published before January 2022. Other relevant papers were selected from the list of references. RESULTS Studies have shown that the complete fertility rate of people living at low altitudes is 7.7, and the complete fertility rate of people living at high altitudes is 4.77, and the hypoxic environment at high altitudes reduces fertility. At the same time, high-altitude, low-oxygen environments are associated with increased infant mortality and post-neonatal mortality. To date, most studies seem to point to a correlation between anoxic exposure at high altitudes and low fertility in humans and animals. CONCLUSION Although the molecular mechanisms are not fully understood, the effects of hypoxia at high altitude on hormonal level, ovarian reserve, embryonic development, testicular development, and sperm motility and levels require further research to investigate this complex topic.
Collapse
Affiliation(s)
- Yan Zhong
- School of Pharmacy, Gansu University of Traditional Chinese Medicine, Lanzhou, China.
- Pharmacy of the 940th Hospital of Joint Logistic Support Force of Chinese People's Liberation Army, Lanzhou, China.
| | - Feifei Liu
- School of Pharmacy, Gansu University of Traditional Chinese Medicine, Lanzhou, China
- Pharmacy of the 940th Hospital of Joint Logistic Support Force of Chinese People's Liberation Army, Lanzhou, China
| | - Xiaojing Zhang
- Pharmacy of the 940th Hospital of Joint Logistic Support Force of Chinese People's Liberation Army, Lanzhou, China.
| | - Qianwen Guo
- Pharmacy of the 940th Hospital of Joint Logistic Support Force of Chinese People's Liberation Army, Lanzhou, China
| | - Zihan Wang
- Pharmacy of the 940th Hospital of Joint Logistic Support Force of Chinese People's Liberation Army, Lanzhou, China
| | - Rong Wang
- Pharmacy of the 940th Hospital of Joint Logistic Support Force of Chinese People's Liberation Army, Lanzhou, China.
| |
Collapse
|
26
|
Liao Z, Tang S, Jiang P, Geng T, Cope DI, Dunn TN, Guner J, Radilla LA, Guan X, Monsivais D. Impaired bone morphogenetic protein (BMP) signaling pathways disrupt decidualization in endometriosis. Commun Biol 2024; 7:227. [PMID: 38402336 PMCID: PMC10894266 DOI: 10.1038/s42003-024-05898-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: 10/20/2023] [Accepted: 02/07/2024] [Indexed: 02/26/2024] Open
Abstract
Endometriosis is linked to increased infertility and pregnancy complications due to defective endometrial decidualization. We hypothesized that identification of altered signaling pathways during decidualization could identify the underlying cause of infertility and pregnancy complications. Our study reveals that transforming growth factor β (TGFβ) pathways are impaired in the endometrium of individuals with endometriosis, leading to defective decidualization. Through detailed transcriptomic analyses, we discovered abnormalities in TGFβ signaling pathways and key regulators, such as SMAD4, in the endometrium of affected individuals. We also observed compromised activity of bone morphogenetic proteins (BMP), a subset of the TGFβ family, that control endometrial receptivity. Using 3-dimensional models of endometrial stromal and epithelial assembloids, we showed that exogenous BMP2 improved decidual marker expression in individuals with endometriosis. Our findings reveal dysfunction of BMP/SMAD signaling in the endometrium of individuals with endometriosis, explaining decidualization defects and subsequent pregnancy complications in these individuals.
Collapse
Affiliation(s)
- Zian Liao
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Graduate Program of Genetics and Genomics, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Suni Tang
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Peixin Jiang
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ting Geng
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Dominique I Cope
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Timothy N Dunn
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, 77030, USA
- Division of Reproductive Endocrinology & Infertility, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Joie Guner
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of Southern California, Los Angeles, CA, 90033, USA
| | - Linda Alpuing Radilla
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xiaoming Guan
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Diana Monsivais
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA.
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
27
|
Liao Z, Tang S, Nozawa K, Shimada K, Ikawa M, Monsivais D, Matzuk MM. Affinity-tagged SMAD1 and SMAD5 mouse lines reveal transcriptional reprogramming mechanisms during early pregnancy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.25.559321. [PMID: 38106095 PMCID: PMC10723262 DOI: 10.1101/2023.09.25.559321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Endometrial decidualization, a prerequisite for successful pregnancies, relies on transcriptional reprogramming driven by progesterone receptor (PR) and bone morphogenetic protein (BMP)-SMAD1/SMAD5 signaling pathways. Despite their critical roles in early pregnancy, how these pathways intersect in reprogramming the endometrium into a receptive state remains unclear. To define how SMAD1 and/or SMAD5 integrate BMP signaling in the uterus during early pregnancy, we generated two novel transgenic mouse lines with affinity tags inserted into the endogenous SMAD1 and SMAD5 loci (Smad1HA/HA and Smad5PA/PA). By profiling the genome-wide distribution of SMAD1, SMAD5, and PR in the mouse uterus, we demonstrated the unique and shared roles of SMAD1 and SMAD5 during the window of implantation. We also showed the presence of a conserved SMAD1, SMAD5, and PR genomic binding signature in the uterus during early pregnancy. To functionally characterize the translational aspects of our findings, we demonstrated that SMAD1/5 knockdown in human endometrial stromal cells suppressed expressions of canonical decidual markers (IGFBP1, PRL, FOXO1) and PR-responsive genes (RORB, KLF15). Here, our studies provide novel tools to study BMP signaling pathways and highlight the fundamental roles of SMAD1/5 in mediating both BMP signaling pathways and the transcriptional response to progesterone (P4) during early pregnancy.
Collapse
Affiliation(s)
- Zian Liao
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Graduate Program of Genetics and Genomics, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Suni Tang
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kaori Nozawa
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Keisuke Shimada
- Research Institute for Microbial Diseases, Osaka University, Osaka, 565-0871, Japan
| | - Masahito Ikawa
- Research Institute for Microbial Diseases, Osaka University, Osaka, 565-0871, Japan
| | - Diana Monsivais
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Martin M. Matzuk
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Graduate Program of Genetics and Genomics, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA
| |
Collapse
|
28
|
Wang B, Li MD, Xu HY, Zhang XC, Bu LG, Li TY, Sun Y, Ni H. Epidermal growth factor: Expression in goat endometrial epithelia during early pregnancy and regulation by interferon tau and FOXO1. Anim Reprod Sci 2024; 261:107406. [PMID: 38141547 DOI: 10.1016/j.anireprosci.2023.107406] [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: 11/07/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023]
Abstract
In ruminants, establishment and maintenance of pregnancy depends upon a well-coordinated interaction between the conceptus and the maternal endometrium. Epidermal growth factor (EGF) is important for embryo implantation and pregnancy establishment. However, the regulatory mechanisms of EGF expression remain unclear. FOXO1, a member of the Forkhead box O (FOXO) subfamily of transcription factors, is currently accepted as a novel endometrial receptivity marker for humans and mice owing to its timely and specific expression at the window of implantation. In this study, we examined the spatiotemporal expression profile of EGF in goat uterus during early pregnancy (Day 0 to Day 50 of pregnancy) and verified that EGF expression was regulated by FOXO1 and interferon tau (IFNT). Our results showed that EGF was highly expressed in the luminal epithelium (LE) and the glandular epithelium (GE) during conceptus adhesion (Day 16 to Day 25 of pregnancy). After implantation, EGF protein signals were continuously detected in the endometrial epithelia and appeared in the conceptus trophectoderm. Furthermore, EGF expression could be up-regulated by IFNT in goat uterus and primary endometrial epithelium cells (EECs). The luciferase assay results showed that FOXO1 could promote EGF transcription by binding to its promoter. And FOXO1 positively regulates EGF expression in goat EECs. These findings contribute to better understanding the role and regulation mechanisms of EGF during ruminant early pregnancy.
Collapse
Affiliation(s)
- Bo Wang
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Meng-Die Li
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Hai-Yue Xu
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Xin-Cheng Zhang
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Li-Ge Bu
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Ting-Yue Li
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Ya Sun
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Hua Ni
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
29
|
Zhang JC, Zhang HL, Xin XY, Zhu YT, Mao X, Hu HQ, Jin YX, Fan RW, Zhang XH, Ye Y, Li D. Mechanisms of Bushen Tiaoxue Granules against controlled ovarian hyperstimulation-induced abnormal morphology of endometrium based on network pharmacology. J Ovarian Res 2024; 17:25. [PMID: 38279186 PMCID: PMC10811918 DOI: 10.1186/s13048-023-01339-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 12/30/2023] [Indexed: 01/28/2024] Open
Abstract
Bushen Tiaoxue Granules (BTG) is an empirical Chinese herbal formula that has been used for the treatment of subfertility. The protective effect of BTG on controlled ovarian hyperstimulation (COH)-induced impaired endometrial receptivity has been reported in our previous study. This study aims to explore the mechanisms of BTG on ameliorating abnormal morphology of endometrium based on network pharmacology. Active compounds of BTG were identified via the traditional Chinese medicine systems pharmacology and UPLC-MS technology. The SwissTargetPrediction platform and HERB database were used to screen out the putative targets of BTG. Potential targets of endometrial dysfunction caused by COH were obtained from three GEO databases. Through the STRING database, the protein-protein interaction was carried out according to the cross-common targets of diseases and drugs. GO terms and KEGG pathways enrichment analyses were conducted via the Metascape database. AutoDock Vina was used for docking validation of the affinity between active compounds and potential targets. Finally, in vivo experiments were used to verify the potential mechanisms derived from network pharmacology study. A total of 141 effective ingredients were obtained from TCMSP and nine of which were verified in UPLC-MS. Six genes were selected through the intersection of 534 disease related genes and 165 drug potential targets. Enrichment analyses showed that BTG might reverse endometrial dysfunction by regulating adherens junction and arachidonic acid metabolism. Hematoxylin-eosin staining revealed that BTG ameliorated the loose and edematous status of endometrial epithelium caused by COH. The protein expression of FOXO1A, β-Catenin and COX-2 was decreased in the COH group, and was up-regulated by BTG. BTG significantly alleviates the edema of endometrial epithelium caused by COH. The mechanisms may be related to adheren junctions and activation of arachidonic acid metabolism. The potential active compounds quercetin, taxifolin, kaempferol, eriodictyol, and isorhamnetin identified from the BTG exhibit marginal cytotoxicity. Both high and low concentrations of kaempferol, eriodictyol, and taxifolin are capable of effectively ameliorating impaired hESC cellular activity.
Collapse
Affiliation(s)
- Jia-Cheng Zhang
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Hao-Lin Zhang
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Xi-Yan Xin
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Yu-Tian Zhu
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Xin Mao
- Department of Radiology, Peking University Third Hospital, Beijing, China
| | - Hang-Qi Hu
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Yu-Xin Jin
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Rui-Wen Fan
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Xiao-Hui Zhang
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Yang Ye
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China.
| | - Dong Li
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
30
|
Diniz WJS, Afonso J, Kertz NC, Dyce PW, Banerjee P. Mapping Expression Quantitative Trait Loci Targeting Candidate Genes for Pregnancy in Beef Cows. Biomolecules 2024; 14:150. [PMID: 38397387 PMCID: PMC10886872 DOI: 10.3390/biom14020150] [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: 12/09/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
Despite collective efforts to understand the complex regulation of reproductive traits, no causative genes and/or mutations have been reported yet. By integrating genomics and transcriptomics data, potential regulatory mechanisms may be unveiled, providing opportunities to dissect the genetic factors governing fertility. Herein, we identified regulatory variants from RNA-Seq data associated with gene expression regulation in the uterine luminal epithelial cells of beef cows. We identified 4676 cis and 7682 trans eQTLs (expression quantitative trait loci) affecting the expression of 1120 and 2503 genes, respectively (FDR < 0.05). These variants affected the expression of transcription factor coding genes (71 cis and 193 trans eQTLs) and genes previously reported as differentially expressed between pregnant and nonpregnant cows. Functional over-representation analysis highlighted pathways related to metabolism, immune response, and hormone signaling (estrogen and GnRH) affected by eQTL-regulated genes (p-value ≤ 0.01). Furthermore, eQTLs were enriched in QTL regions for 13 reproduction-related traits from the CattleQTLdb (FDR ≤ 0.05). Our study provides novel insights into the genetic basis of reproductive processes in cattle. The underlying causal mechanisms modulating the expression of uterine genes warrant further investigation.
Collapse
Affiliation(s)
- Wellison J. S. Diniz
- Department of Animal Sciences, Auburn University, Auburn, AL 36849, USA; (N.C.K.); (P.W.D.); (P.B.)
| | - Juliana Afonso
- Embrapa Pecuária Sudeste, Rodovia Washington Luiz, Km 234, s/n, Fazenda Canchim, São Carlos 13560-970, SP, Brazil;
| | - Nicholas C. Kertz
- Department of Animal Sciences, Auburn University, Auburn, AL 36849, USA; (N.C.K.); (P.W.D.); (P.B.)
| | - Paul W. Dyce
- Department of Animal Sciences, Auburn University, Auburn, AL 36849, USA; (N.C.K.); (P.W.D.); (P.B.)
| | - Priyanka Banerjee
- Department of Animal Sciences, Auburn University, Auburn, AL 36849, USA; (N.C.K.); (P.W.D.); (P.B.)
| |
Collapse
|
31
|
Wu S, Wu G, Li Y, Wu H. Mechanism of HCG regulating epithelial-mesenchymal transition progression in endometrial cells through the FoxO1/miR223-5p/Wnt5α pathway. Reprod Biomed Online 2024; 48:103246. [PMID: 37903673 DOI: 10.1016/j.rbmo.2023.05.016] [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: 02/06/2023] [Revised: 05/22/2023] [Accepted: 05/25/2023] [Indexed: 11/01/2023]
Abstract
RESEARCH QUESTION Does human chorionic gonadotrophin (HCG) influence endometrial receptivity and epithelial-mesenchymal transition (EMT) via the FoxO1/miR223-5p/Wnt5α pathway? DESIGN This study aimed to establish the co-culture system of human embryonic trophoblast cell line (HTR-8-Svneo) cells and human endometrial epithelial cell line (HEEC) cells. The expression of Wnt5α protein and EMT-related proteins in HTR-8-Svneo and HEEC cells treated in a gradient-dependent manner using HCG and exosome inhibitor GW4869 were detected in the co-culture system. RESULTS In the HTR-8-Svneo/HEEC co-culture system, miR223-5p in HEEC cells increased significantly with induction of HTR-8-Svneo cells by 100 IU/ml HCG for 48 h (P = 0.046), and Wnt5α protein decreased significantly in HEEC cells (P = 0.021). Pretreatment of HTR-8-Svneo cells with GW4869, and knockdown of FoxO1 in HTR-8-Svneo cells, significantly inhibited the above effects of HCG on miR223-5p and Wnt5α expression in HEEC cells in the HTR-8-Svneo/HEEC co-culture system. HTR-8-Svneo cells induced with 100 IU/ml HCG for 48 h significantly enhanced the logarithmic phase proliferation activity of HEEC cells in the co-culture system (P < 0.001), while knockdown of FoxO1 in HTR-8-Svneo cells and inhibition of miR223-5p in HEEC cells suppressed proliferation of HEEC cells in the HTR-8-Svneo/HEEC co-culture system (P < 0.001). CONCLUSIONS HCG exposure induces HTR-8-Svneo cells to up-regulate miR223-5p expression, which enters HEEC cells in the co-culture system through the exosomal pathway, and inhibits Wnt5α expression and the progress of EMT.
Collapse
Affiliation(s)
- Shuxie Wu
- Obstetrics and Gynaecology Hospital of Fudan University, Shanghai, China
| | - Gao Wu
- Department of Pharmacy, First Affiliated Hospital of Naval Military Medical University, Shanghai, China
| | - Yanping Li
- Reproductive Medicine Centre, Xiangya Hospital, Central South University, Hunan, China.
| | - Hanbin Wu
- Clinical Pharmacy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
32
|
Monsivais D, Liao Z, Tang S, Jiang P, Geng T, Cope D, Dunn T, Guner J, Radilla LA, Guan X. Impaired bone morphogenetic protein (BMP) signaling pathways disrupt decidualization in endometriosis. RESEARCH SQUARE 2023:rs.3.rs-3471243. [PMID: 37986901 PMCID: PMC10659538 DOI: 10.21203/rs.3.rs-3471243/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Endometriosis is linked to increased infertility and pregnancy complications due to defective endometrial decidualization. We hypothesized that identification of altered signaling pathways during decidualization could identify the underlying cause of infertility and pregnancy complications. Our study reveals that transforming growth factor β (TGFβ) pathways are impaired in the endometrium of individuals with endometriosis, leading to defective decidualization. Through detailed transcriptomic analyses, we discovered abnormalities in TGFβ signaling pathways and key regulators, such as SMAD4, in the endometrium of affected individuals. We also observed compromised activity of bone morphogenetic proteins (BMP), a subset of the TGFβ family, that control endometrial receptivity. Using 3-dimensional models of endometrial stromal and epithelial assembloids, we showed that exogenous BMP2 improved decidual marker expression in individuals with endometriosis. Our findings unveil a previously unidentified dysfunction in BMP/SMAD signaling in the endometrium of individuals with endometriosis, explaining decidualization defects and subsequent pregnancy complications in these individuals.
Collapse
|
33
|
Marquardt RM, Tran DN, Lessey BA, Rahman MS, Jeong JW. Epigenetic Dysregulation in Endometriosis: Implications for Pathophysiology and Therapeutics. Endocr Rev 2023; 44:1074-1095. [PMID: 37409951 PMCID: PMC10638603 DOI: 10.1210/endrev/bnad020] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 02/15/2023] [Accepted: 07/05/2023] [Indexed: 07/07/2023]
Abstract
Endometriosis is a prevalent gynecological condition associated with pelvic pain and infertility. Despite more than a century of research, the etiology of endometriosis still eludes scientific consensus. This lack of clarity has resulted in suboptimal prevention, diagnosis, and treatment options. Evidence of genetic contributors to endometriosis is interesting but limited; however, significant progress has been made in recent years in identifying an epigenetic role in the pathogenesis of endometriosis through clinical studies, in vitro cell culture experiments, and in vivo animal models. The predominant findings include endometriosis-related differential expression of DNA methyltransferases and demethylases, histone deacetylases, methyltransferases, and demethylases, and regulators of chromatin architecture. There is also an emerging role for miRNAs in controlling epigenetic regulators in the endometrium and endometriosis. Changes in these epigenetic regulators result in differential chromatin organization and DNA methylation, with consequences for gene expression independent of a genetic sequence. Epigenetically altered expression of genes related to steroid hormone production and signaling, immune regulation, and endometrial cell identity and function have all been identified and appear to play into the pathophysiological mechanisms of endometriosis and resulting infertility. This review summarizes and critically discusses early seminal findings, the ever-growing recent evidence of epigenetic contributions to the pathophysiology of endometriosis, and implications for proposed epigenetically targeted therapeutics.
Collapse
Affiliation(s)
- Ryan M Marquardt
- Department of Obstetrics, Gynecology & Reproductive Biology, Michigan State University, Grand Rapids, MI, USA
| | - Dinh Nam Tran
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, USA
| | - Bruce A Lessey
- Department of Obstetrics and Gynecology, Wake Forest Baptist Health, Winston-Salem, NC, USA
| | - Md Saidur Rahman
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, USA
| | - Jae-Wook Jeong
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, USA
| |
Collapse
|
34
|
Bu LG, Wang B, Li TY, Sun Y, Kong LL, Zhao ZA, Li SJ, Ding NZ, Ni H. An IFNT/FOXO1/PTGS2 axis regulates prostaglandin F 2α synthesis in goat uterus during early pregnancy. J Dairy Sci 2023; 106:8060-8071. [PMID: 37268579 DOI: 10.3168/jds.2022-23153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 04/25/2023] [Indexed: 06/04/2023]
Abstract
In ruminants, IFN-tau (IFNT) regulates the production of prostaglandins (PG) in the endometrium, which is crucial for conceptus adhesion. However, the related molecular regulatory mechanisms remain unclear. Forkhead box O1 (FOXO1), a member of the FOXO subfamily of transcription factors, is known to be important for mouse implantation and decidualization. In this study, we determined the spatiotemporal expression profile of FOXO1 in goat endometrium during early pregnancy. FOXO1 was highly expressed in the glandular epithelium since the onset of conceptus adhesion (d 16 of pregnancy). Then, we validated that FOXO1 could bind to the promoter of prostaglandin-endoperoxide synthase 2 (PTGS2) and increase its transcription. And the expression profile of PTGS2 was similar to that of FOXO1 in the peri-implantation uterus. Moreover, IFNT could upregulate the levels of FOXO1 and PTGS2 in goat uterus and primary endometrial epithelium cells (EEC). In EEC, the intracellular content of PGF2α was positively correlated with the levels of IFNT and FOXO1. Altogether, we found an IFNT/FOXO1/PTGS2 axis that controls the synthesis of PGF2α but not prostaglandin E2 in goat uterine glands. These findings contribute to better understanding the function of FOXO1 in the reproductive physiology of goats and provide more insights into the implantation of small ruminants.
Collapse
Affiliation(s)
- Li-Ge Bu
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Bo Wang
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Ting-Yue Li
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Ya Sun
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Li-Li Kong
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Zhen-Ao Zhao
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Shi-Jie Li
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Nai-Zheng Ding
- Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Science, Shandong Normal University, Jinan 250014, China
| | - Hua Ni
- Key Laboratory of Animal Cellular and Genetic Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
35
|
de Jesus Nascimento AE, Santos LC, Santos BR, Santos EO, Cunha MCDSG, Snoeck PPDN, de Lavor MSL, Silva JF. Spatial and temporal expression profile of sex steroid receptors and antioxidant enzymes in the maternal-fetal interface of domestic cats. Theriogenology 2023; 210:234-243. [PMID: 37542738 DOI: 10.1016/j.theriogenology.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/07/2023]
Abstract
Sex steroids and antioxidant enzymes modulate uterine and placental physiology. Failures in the expression, signaling, and/or secretion of these mediators are associated with female infertility and gestational problems. However, there is no data on the expression profile of receptors for sex steroids and antioxidant enzymes in the maternal-fetal interface of domestic cats. Uterus and placenta samples from non-pregnant diestrus cats and cats in mid- and late pregnancy were used to analyze the protein and gene expression of the receptors for estrogen alpha (ERα), progesterone (PR), and androgen (AR) and the antioxidant enzymes superoxide dismutase 1 (SOD1), catalase, and glutathione peroxidase 1 (GPX1) by immunohistochemistry and qPCR. Higher uterine expression of ERα, Pr, and Sod1 was observed in the pregnant cats, especially in mid-pregnancy, compared to non-pregnant diestrus cats, as well as reduced endometrial catalase immunostaining. In the placenta, the mRNA expression of Erα, Pr, Ar, and Gpx1 was higher in late pregnancy in relation to mid-pregnancy. Moreover, weak or no placental expression was observed for catalase in mid- and late pregnancy, while strong immunostaining was observed for AR in trophoblasts and decidual cells in mid-pregnancy. The findings of this study demonstrated that pregnancy in female cats increases the uterine expression of sex steroid receptors and antioxidant enzymes, and that the placental expression of these mediators varies according to gestational age.
Collapse
Affiliation(s)
- Acácia Eduarda de Jesus Nascimento
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Luciano Cardoso Santos
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Bianca Reis Santos
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Emilly Oliveira Santos
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Maria Clara da Silva Galrão Cunha
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Paola Pereira das Neves Snoeck
- Hospital Veterinário, Departamento de Ciências Agrarias e Ambientais, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Mário Sérgio Lima de Lavor
- Hospital Veterinário, Departamento de Ciências Agrarias e Ambientais, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil
| | - Juneo Freitas Silva
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilhéus, Brazil.
| |
Collapse
|
36
|
Liao Z, Tang S, Jiang P, Geng T, Cope DI, Dunn TN, Guner J, Radilla LA, Guan X, Monsivais D. Impaired bone morphogenetic protein signaling pathways disrupt decidualization in endometriosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.21.558268. [PMID: 37790548 PMCID: PMC10542516 DOI: 10.1101/2023.09.21.558268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
It is hypothesized that impaired endometrial decidualization contributes to decreased fertility in individuals with endometriosis. To identify the molecular defects that underpin defective decidualization in endometriosis, we subjected endometrial stromal cells from individuals with or without endometriosis to time course in vitro decidualization with estradiol, progesterone, and 8-bromo-cyclic-AMP (EPC) for 2, 4, 6, or 8 days. Transcriptomic profiling identified differences in key pathways between the two groups, including defective bone morphogenetic protein (BMP)/SMAD4 signaling (ID2, ID3, FST), oxidate stress response (NFE2L2, ALOX15, SLC40A1), and retinoic acid signaling pathways (RARRES, RARB, ALDH1B1). Genome-wide binding analyses identified an altered genomic distribution of SMAD4 and H3K27Ac in the decidualized stromal cells from individuals without endometriosis relative to those with endometriosis, with target genes enriched in pathways related to signaling by transforming growth factor β (TGFβ), neurotrophic tyrosine kinase receptors (NTRK), and nerve growth factor (NGF)-stimulated transcription. We found that direct SMAD1/5/4 target genes control FOXO, PI3K/AKT, and progesterone-mediated signaling in decidualizing cells and that BMP2 supplementation in endometriosis patient-derived assembloids elevated the expression of decidualization markers. In summary, transcriptomic and genome-wide binding analyses of patient-derived endometrial cells and assembloids identified that a functional BMP/SMAD1/5/4 signaling program is crucial for engaging decidualization.
Collapse
Affiliation(s)
- Zian Liao
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Graduate Program of Genetics and Genomics, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Suni Tang
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Peixin Jiang
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Thoracic/Head and Neck Medical Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ting Geng
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Dominique I. Cope
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Timothy N. Dunn
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, 77030, USA
- Division of Reproductive Endocrinology & Infertility, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Joie Guner
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of Southern California, Los Angeles, CA, 90033, USA
| | - Linda Alpuing Radilla
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xiaoming Guan
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Diana Monsivais
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX, 77030, USA
| |
Collapse
|
37
|
Jin S, Wang TT, Huang JC, Wang YS, Guo B, Yue ZP. Melatonin modulates endometrial decidualization via NOTCH1-NRF2-FOXO1-GSH pathway†. Biol Reprod 2023; 109:299-308. [PMID: 37334936 DOI: 10.1093/biolre/ioad066] [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: 04/03/2023] [Revised: 05/19/2023] [Accepted: 06/11/2023] [Indexed: 06/21/2023] Open
Abstract
Melatonin is important for oocyte maturation, fertilization, early embryonic development, and embryo implantation, but less knowledge is available regarding its role in decidualization. The present study found that melatonin did not alter the proliferation of human endometrial stromal cells (ESCs), as well as cell cycle progress, but suppressed stromal differentiation after binding to the melatonin receptor 1B (MTNR1B), which was visualized in decidualizing ESCs. Further analysis evidenced that application of melatonin resulted in the diminishment for NOTCH1 and RBPJ expression. Supplementation of recombinant NOTCH1 protein (rNOTCH1) counteracted the impairment of stromal differentiation conferred by melatonin, while the addition of the NOTCH signaling pathway inhibitor DAPT aggravated the differentiation progress. Meanwhile, melatonin might restrain the expression and transcriptional activity of nuclear factor erythroid 2-related factor 2 (NRF2), whose blockage accelerated the fault of stromal differentiation under the context of melatonin, but this restraint was subsequently ameliorated by rNOTCH1. Forkhead box O 1 (FOXO1) was identified as a downstream target of melatonin in decidualization. Repression of NRF2 antagonized the retrieval of rNOTCH1 due to aberrant FOXO1 expression elicited by melatonin. Moreover, melatonin brought about the occurrence of oxidative stress accompanied by an obvious accumulation of intracellular reactive oxygen species and a significant reduction in glutathione (GSH) content, as well as enzymatic activities of glutathione peroxidase and glutathione reductase, whereas supplementation of rNOTCH1 improved the above-mentioned effects. Nevertheless, this improvement was disrupted by the blockage of NRF2 and FOXO1. Furthermore, addition of GSH rescued the defect of stromal differentiation by melatonin. Collectively, melatonin might impair endometrial decidualization by restraining the differentiation of ESCs dependent on NOTCH1-NRF2-FOXO1-GSH pathway after binding to the MTNR1B receptor.
Collapse
Affiliation(s)
- Shan Jin
- College of Veterinary Medicine, Jilin University, Changchun, P. R. China
- Reproductive Medical Center, the Second Hospital of Jilin University, Changchun, P. R. China
| | - Ting-Ting Wang
- College of Veterinary Medicine, Jilin University, Changchun, P. R. China
| | - Ji-Cheng Huang
- College of Veterinary Medicine, Jilin University, Changchun, P. R. China
| | - Yu-Si Wang
- College of Veterinary Medicine, Jilin University, Changchun, P. R. China
| | - Bin Guo
- College of Veterinary Medicine, Jilin University, Changchun, P. R. China
| | - Zhan-Peng Yue
- College of Veterinary Medicine, Jilin University, Changchun, P. R. China
| |
Collapse
|
38
|
Guo J, Zhou W, Sacco M, Downing P, Dimitriadis E, Zhao F. Using organoids to investigate human endometrial receptivity. Front Endocrinol (Lausanne) 2023; 14:1158515. [PMID: 37693361 PMCID: PMC10484744 DOI: 10.3389/fendo.2023.1158515] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 04/13/2023] [Indexed: 09/12/2023] Open
Abstract
The human endometrium is only receptive to an implanting blastocyst in the mid-secretory phase of each menstrual cycle. Such time-dependent alterations in function require intricate interplay of various factors, largely coordinated by estrogen and progesterone. Abnormal endometrial receptivity is thought to contribute to two-thirds of the implantation failure in humans and therefore significantly hindering IVF success. Despite the incontrovertible importance of endometrial receptivity in implantation, the precise mechanisms involved in the regulation of endometrial receptivity remain poorly defined. This is mainly due to a lack of proper in vitro models that recapitulate the in vivo environment of the receptive human endometrium. Organoids were recently established from human endometrium with promising features to better mimic the receptive phase. Endometrial organoids show long-term expandability and the capability to preserve the structural and functional characteristics of the endometrial tissue of origin. This three-dimensional model maintains a good responsiveness to steroid hormones in vitro and replicates key morphological features of the receptive endometrium in vivo, including pinopodes and pseudostratified epithelium. Here, we review the current findings of endometrial organoid studies that have been focused on investigating endometrial receptivity and place an emphasis on methods to further refine and improve this model.
Collapse
Affiliation(s)
- Junhan Guo
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Zhou
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC, Australia
- Gynaecology Research Centre, Royal Women’s Hospital, Parkville, VIC, Australia
| | - Michaela Sacco
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC, Australia
- Gynaecology Research Centre, Royal Women’s Hospital, Parkville, VIC, Australia
| | - Poppy Downing
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC, Australia
- Gynaecology Research Centre, Royal Women’s Hospital, Parkville, VIC, Australia
| | - Evdokia Dimitriadis
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC, Australia
- Gynaecology Research Centre, Royal Women’s Hospital, Parkville, VIC, Australia
| | - Feifei Zhao
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
39
|
Ye Q, Li H, Xu B, He Z, Yan X. Butyrate Improves Porcine Endometrial Epithelial Cell Receptivity via Enhancing Acetylation of Histone H3K9. Mol Nutr Food Res 2023; 67:e2200703. [PMID: 37417211 DOI: 10.1002/mnfr.202200703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 04/26/2023] [Indexed: 07/08/2023]
Abstract
SCOPE Uterine receptivity is a major restriction of embryo implantation and survival, and the endometrial luminal epithelium serves as the transient gateway for uterine receptivity and embryo implantation. Butyrate is reported to promote the success of embryo implantation, but the effects and mechanism of butyrate on uterine receptivity are still unknown. METHODS AND RESULTS Porcine endometrial epithelial cells (PEECs) are used as a model, and the cellular receptivity changes, metabolism, and gene expression profiles influenced by butyrate are analyzed. The study finds that butyrate improves receptive changes in PEECs, including inhibiting proliferation, exhibiting more pinocytosis on the cell surface, and increasing adhesiveness to porcine trophoblast cells. In addition, butyrate increases prostaglandin synthesis and markedly impacts purine metabolism, pyrimidine metabolism, and the FoxO signaling pathway. siRNA to inhibit the expression of FoxO1 and chromatin immunoprecipitation-sequencing (ChIP-seq) of H3K9ac are used to demonstrate that the H3K9ac/FoxO1/PCNA pathway can contribute to the effects of cell proliferation inhibition and uterine receptivity improvement induced by butyrate. CONCLUSION The findings reveal that butyrate improves endometrial epithelial cell receptivity by enhancing the acetylation of histone H3K9, which shows nutritional regulation and therapeutic potential for poor uterine receptivity and difficulty in embryo implantation.
Collapse
Affiliation(s)
- Qianhong Ye
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal, Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, China
- Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, Hubei, 430070, China
| | - Haoyu Li
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal, Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, China
- Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, Hubei, 430070, China
| | - Baoyang Xu
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal, Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, China
- Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, Hubei, 430070, China
| | - Ziyi He
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal, Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, China
- Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, Hubei, 430070, China
| | - Xianghua Yan
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Frontiers Science Center for Animal, Breeding and Sustainable Production, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, China
- Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, Hubei, 430070, China
| |
Collapse
|
40
|
METTL3 is essential for normal progesterone signaling during embryo implantation via m 6A-mediated translation control of progesterone receptor. Proc Natl Acad Sci U S A 2023; 120:e2214684120. [PMID: 36693099 PMCID: PMC9945998 DOI: 10.1073/pnas.2214684120] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Embryo implantation, a crucial step in human reproduction, is tightly controlled by estrogen and progesterone (P4) via estrogen receptor alpha and progesterone receptor (PGR), respectively. Here, we report that N6-methyladenosine (m6A), the most abundant mRNA modification in eukaryotes, plays an essential role in embryo implantation through the maintenance of P4 signaling. Conditional deletion of methyltransferase-like 3 (Mettl3), encoding the m6A writer METTL3, in the female reproductive tract using a Cre mouse line with Pgr promoter (Pgr-Cre) resulted in complete implantation failure due to pre-implantation embryo loss and defective uterine receptivity. Moreover, the uterus of Mettl3 null mice failed to respond to artificial decidualization. We further found that Mettl3 deletion was accompanied by a marked decrease in PGR protein expression. Mechanistically, we found that Pgr mRNA is a direct target for METTL3-mediated m6A modification. A luciferase assay revealed that the m6A modification in the 5' untranslated region (5'-UTR) of Pgr mRNA enhances PGR protein translation efficiency in a YTHDF1-dependent manner. Finally, we demonstrated that METTL3 is required for human endometrial stromal cell decidualization in vitro and that the METTL3-PGR axis is conserved between mice and humans. In summary, this study provides evidence that METTL3 is essential for normal P4 signaling during embryo implantation via m6A-mediated translation control of Pgr mRNA.
Collapse
|
41
|
Integrating Analysis to Identify Differential circRNAs Involved in Goat Endometrial Receptivity. Int J Mol Sci 2023; 24:ijms24021531. [PMID: 36675045 PMCID: PMC9865150 DOI: 10.3390/ijms24021531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/27/2022] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
Endometrial receptivity is one of the main factors underlying a successful pregnancy, with reports substantiating the fact that suboptimal endometrial receptivity accounts for two-thirds of early implantation event failures. The association between circRNAs and endometrial receptivity in the goat remains unclear. This study aims to identify potential circRNAs and regulatory mechanisms related to goat endometrial receptivity. Therefore, the endometrial samples on day 16 of pregnancy and day 16 of the estrous cycle were analyzed using high-throughput RNA-seq and bioinformatics. The results show that 4666 circRNAs were identified, including 7 downregulated and 11 upregulated differentially expressed circRNAs (DE-circRNAs). Back-splicing and RNase R resistance verified the identified circRNAs. We predicted the competing endogenous RNA (ceRNA) regulatory mechanism and potential target genes of DE-circRNAs. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses of these predicted target genes suggest that DE-circRNAs were significantly involved in establishing endometrial receptivity. Furthermore, Sanger sequencing, qPCR, correlation analysis and Fluorescence in Situ Hybridization (FISH) show that circ_MYRF derived from the host gene myelin regulatory factor (MYRF) might regulate the expression of interferon stimulating gene 15 (ISG15), thereby promoting the formation of endometrial receptivity. These novel findings may contribute to a better understanding of the molecular mechanisms regulating endometrial receptivity and promoting the maternal recognition of pregnancy (MRP).
Collapse
|
42
|
Xie K, Li Y, He G, Zhao X, Chen D, Yu B, Luo Y, Mao X, Huang Z, Yu J, Luo J, Zheng P, Yan H, Li H, He J. Daidzein supplementation improved fecundity in sows via modulation of ovarian oxidative stress and inflammation. J Nutr Biochem 2022; 110:109145. [PMID: 36049671 DOI: 10.1016/j.jnutbio.2022.109145] [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: 03/18/2021] [Revised: 10/15/2021] [Accepted: 08/10/2022] [Indexed: 01/13/2023]
Abstract
Adequate ovarian hormones secretion is essential for pregnancy success. Oxidative damage and following inflammation can destroy the ovarian normal function in mammals. Daidzein (DAI) is a classical isoflavonic phytoestrogen with specific oestrogenic activity. This study aimed to explore the effects of daidzein supplementation on fertility and ovarian characteristics of sows through biochemical analysis and RNA-seq technology. Twelve multiparous Yorkshire × Landrace sows were randomly divided into CON and DAI groups. We found that DAI increased total number of embryos as well as P4 and E2 levels of serum. DAI not only elevated the activities of T-AOC and GSH-Px, but also tended to decrease the content of MDA and IL-6 in the serum. In ovary, RNA-Seq identified 237 differentially expressed genes (DEGs), and GO analysis showed that these DEGs were linked to functions associated with immune dysfunction. Moreover, STRING analysis demonstrated that most interacting nodes were TLR-4, LCP2, and CD86. Furthermore, DAI decreased the content of MDA, IL-1β, IL-6, and TNF-α, and increased the activities of T-AOC and CAT in ovarian tissue. Interestingly, a partial mantel correlation showed that T-AOC was the strongest correlation between the ovarian dataset and selected DEGs. Additionally, DAI supplementation not only increased the protein expressions of Nrf2, HO-1, and NQO1, but also decreased the protein expressions of TLR-4, p-NFκB, p-AKT, and p-IκBα. Altogether, our results indicated that DAI could ameliorate ovarian oxidative stress and inflammation in sows, which might be mediated by suppressing the TLR4/NF-κB signaling pathway and activating the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Kunhong Xie
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China; Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan, P. R. China
| | - Yan Li
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China; Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan, P. R. China
| | - Guoru He
- New Hope Liuhe Co., Ltd. Sichuan Province, Chengdu, Sichuan, P. R. China
| | - Xuefeng Zhao
- Shandong Animal Product Quality and Safety Center, Jinan, Shangdong, P. R. China
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China; Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan, P. R. China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China; Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan, P. R. China
| | - Yuheng Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China; Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan, P. R. China.
| | - Xiangbing Mao
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China; Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan, P. R. China
| | - Zhiqing Huang
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China; Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan, P. R. China
| | - Jie Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China; Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan, P. R. China
| | - Junqiu Luo
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China; Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan, P. R. China
| | - Ping Zheng
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China; Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan, P. R. China
| | - Hui Yan
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China; Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan, P. R. China
| | - Hua Li
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China; Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan, P. R. China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, Sichuan, P. R. China; Key Laboratory of Animal Disease-resistant Nutrition, Chengdu, Sichuan, P. R. China.
| |
Collapse
|
43
|
Lee SH, Lim CL, Shen W, Tan SMX, Woo ARE, Yap YHY, Sian CAS, Goh WWB, Yu WP, Li L, Lin VCL. Activation function 1 of progesterone receptor is required for progesterone antagonism of oestrogen action in the uterus. BMC Biol 2022; 20:222. [PMID: 36199058 PMCID: PMC9535881 DOI: 10.1186/s12915-022-01410-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 09/13/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Progesterone receptor (PGR) is a master regulator of uterine function through antagonistic and synergistic interplays with oestrogen receptors. PGR action is primarily mediated by activation functions AF1 and AF2, but their physiological significance is unknown. RESULTS We report the first study of AF1 function in mice. The AF1 mutant mice are infertile with impaired implantation and decidualization. This is associated with a delay in the cessation of epithelial proliferation and in the initiation of stromal proliferation at preimplantation. Despite tissue selective effect on PGR target genes, AF1 mutations caused global loss of the antioestrogenic activity of progesterone in both pregnant and ovariectomized models. Importantly, the study provides evidence that PGR can exert an antioestrogenic effect by genomic inhibition of Esr1 and Greb1 expression. ChIP-Seq data mining reveals intermingled PGR and ESR1 binding on Esr1 and Greb1 gene enhancers. Chromatin conformation analysis shows reduced interactions in these genes' loci in the mutant, coinciding with their upregulations. CONCLUSION AF1 mediates genomic inhibition of ESR1 action globally whilst it also has tissue-selective effect on PGR target genes.
Collapse
Affiliation(s)
- Shi Hao Lee
- grid.59025.3b0000 0001 2224 0361School of Biological Sciences, Nanyang Technological University, Singapore, 637551 Singapore
| | - Chew Leng Lim
- grid.59025.3b0000 0001 2224 0361School of Biological Sciences, Nanyang Technological University, Singapore, 637551 Singapore
| | - Wei Shen
- grid.35155.370000 0004 1790 4137College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Samuel Ming Xuan Tan
- grid.59025.3b0000 0001 2224 0361School of Biological Sciences, Nanyang Technological University, Singapore, 637551 Singapore
| | - Amanda Rui En Woo
- grid.59025.3b0000 0001 2224 0361School of Biological Sciences, Nanyang Technological University, Singapore, 637551 Singapore
| | - Yeannie H. Y. Yap
- grid.59025.3b0000 0001 2224 0361School of Biological Sciences, Nanyang Technological University, Singapore, 637551 Singapore ,grid.459705.a0000 0004 0366 8575Present Address: Department of Oral Biology and Biomedical Sciences, Faculty of Dentistry, MAHSA University, Bandar Saujana Putra, 42610 Jenjarom, Selangor Malaysia
| | - Caitlyn Ang Su Sian
- grid.59025.3b0000 0001 2224 0361School of Biological Sciences, Nanyang Technological University, Singapore, 637551 Singapore
| | - Wilson Wen Bin Goh
- grid.59025.3b0000 0001 2224 0361School of Biological Sciences, Nanyang Technological University, Singapore, 637551 Singapore
| | - Wei-Ping Yu
- grid.185448.40000 0004 0637 0221Animal Gene Editing Laboratory (AGEL), Biological Resource Centre, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673 Singapore ,grid.418812.60000 0004 0620 9243Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673 Singapore
| | - Li Li
- College of Informatics, Huazhong Agricultural University, Wuhan, China.
| | - Valerie C. L. Lin
- grid.59025.3b0000 0001 2224 0361School of Biological Sciences, Nanyang Technological University, Singapore, 637551 Singapore
| |
Collapse
|
44
|
Hewitt SC, Wu SP, Wang T, Ray M, Brolinson M, Young SL, Spencer TE, DeCherney A, DeMayo FJ. The Estrogen Receptor α Cistrome in Human Endometrium and Epithelial Organoids. Endocrinology 2022; 163:bqac116. [PMID: 35895287 PMCID: PMC9368022 DOI: 10.1210/endocr/bqac116] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Indexed: 11/19/2022]
Abstract
Endometrial health is affected by molecular processes that underlie estrogen responses. We assessed estrogen regulation of endometrial function by integrating the estrogen receptor α (ESR1) cistromes and transcriptomes of endometrial biopsies taken from the proliferative and mid-secretory phases of the menstrual cycle together with hormonally stimulated endometrial epithelial organoids. The cycle stage-specific ESR1 binding sites were determined by chromatin immunoprecipitation and next-generation sequencing and then integrated with changes in gene expression from RNA sequencing data to infer candidate ESR1 targets in normal endometrium. Genes with ESR1 binding in whole endometrium were enriched for chromatin modification and regulation of cell proliferation. The distribution of ESR1 binding sites in organoids was more distal from gene promoters when compared to primary endometrium and was more similar to the proliferative than the mid-secretory phase ESR1 cistrome. Inferred organoid estrogen/ESR1 candidate target genes affected formation of cellular protrusions and chromatin modification. Comparison of signaling effected by candidate ESR1 target genes in endometrium vs organoids reveals enrichment of both overlapping and distinct responses. Our analysis of the ESR1 cistromes and transcriptomes from endometrium and organoids provides important resources for understanding how estrogen affects endometrial health and function.
Collapse
Affiliation(s)
- Sylvia C Hewitt
- Pregnancy & Female Reproduction, RDBL, NIEHS, Research Triangle Park, North Carolina 27709, USA
| | - San-pin Wu
- Pregnancy & Female Reproduction, RDBL, NIEHS, Research Triangle Park, North Carolina 27709, USA
| | - Tianyuan Wang
- Integrative Bioinformatics Support Group, NIEHS, Research Triangle Park, North Carolina 27709, USA
| | - Madhumita Ray
- Pregnancy & Female Reproduction, RDBL, NIEHS, Research Triangle Park, North Carolina 27709, USA
| | - Marja Brolinson
- Program in Reproductive and Adult Endocrinology, NICHD, Bethesda, Maryland 20847, USA
| | - Steven L Young
- Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | - Thomas E Spencer
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri, Columbia, Missouri 65211, USA
| | - Alan DeCherney
- Program in Reproductive and Adult Endocrinology, NICHD, Bethesda, Maryland 20847, USA
| | - Francesco J DeMayo
- Pregnancy & Female Reproduction, RDBL, NIEHS, Research Triangle Park, North Carolina 27709, USA
| |
Collapse
|
45
|
Das M, Mohanty SR, Minocha T, Mishra NK, Yadav SK, Haldar C. Circadian desynchronization in pregnancy of Golden hamster following long time light exposure: Involvement of Akt/FoxO1 pathway. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2022; 234:112508. [PMID: 35841738 DOI: 10.1016/j.jphotobiol.2022.112508] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 06/18/2022] [Accepted: 06/27/2022] [Indexed: 06/15/2023]
Abstract
Coordination between central and peripheral reproductive clocks in females is poorly understood. Long light is having a hazardous effect on reproductive health. Hence, explored the effect of long-time light exposure (LLD; 16L:8D) on the central and peripheral reproductive (ovary and uterus) clock genes (Bmal1, Clock, Per1, Per2, Cry1 and Cry2) and its downstream regulators (Aanat, Egf, Cx26, Cx43, ERα, pAktS-473, pAktT-308, pFoxO1T-24, 14-3-3, HoxA10, HoxA11 and Pibf) expression in non-pregnant and pregnant Golden hamster. Young adult Golden hamsters were exposed to LLD for 30 days and then were mated. We observed that LLD exposure increased the thickness of the endometrium and reduced myometrium thickness, resembling uterine adenomyosis. In non-pregnant females LLD altered the expressions of clock genes in suprachiasmatic nuclei (SCN), ovary and the uterus along with serum estradiol rhythm. LLD upregulated Egf and downregulated Aanat, Cx26, and Cx43 mRNA levels in uterus. LLD upregulated Akt/FoxO1 phosphorylation and 14-3-3 expressions in the uterus of nonpregnant females. LLD exposure to pregnant females lowered serum progesterone, Aanat, Pibf, Hoxa10, and Hoxa11 mRNA expressions on D4 (peri-implantation) and D8 (post-implantation) resulting in a low implantation rate on D8 (post-implantation). Hence it is evident that the frequent pregnancy anomalies noted under a long light schedule might be due to desynchronization in Aanat, Pibf, Hoxa10, and Hoxa11 as well as the central and peripheral clock genes (Bmal1, Clock, Per1, Per2, Cry1 and Cry2). LLD exposure desynchronized the central and peripheral reproductive clock affecting uterine physiology via Akt/FoxO1 pathway in Golden hamsters. Thus, LLD is a risk factor for female reproductive health and fertility.
Collapse
Affiliation(s)
- Megha Das
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Soumya Ranjan Mohanty
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Tarun Minocha
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Nitesh Kumar Mishra
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Sanjeev Kumar Yadav
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India.
| | - Chandana Haldar
- Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
46
|
Aberrant gene expression of superoxide dismutases in Chlamydia trachomatis-infected recurrent spontaneous aborters. Sci Rep 2022; 12:14688. [PMID: 36038649 PMCID: PMC9424283 DOI: 10.1038/s41598-022-18941-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 08/22/2022] [Indexed: 11/19/2022] Open
Abstract
Study aimed to characterize the expression of antioxidant genes SOD1 and SOD2 in Chlamydia trachomatis-induced recurrent spontaneous aborters and further determine their role by in silico analysis. First void urine was collected from 130 non-pregnant women with history of recurrent spontaneous abortion (RSA) (Group I) and 130 non-pregnant women (Group II; control) attending Obstetrics and Gynecology Department, SJH, New Delhi, India. C. trachomatis detection was performed by conventional PCR in urine. Gene expression of SOD1 and SOD2 was performed by quantitative real-time PCR. Further, its interacting partners were studied by in silico analysis. 22 patients were positive for C. trachomatis in Group I. Significant upregulation was observed for SOD2 gene in C. trachomatis-infected RSA patients while SOD1 was found to be downregulated. Increased concentration of oxidative stress biomarkers 8-hydroxyguanosine and 8-isoprostane was found in C. trachomatis-infected RSA patients. Protein–protein interaction (PPI) of SOD proteins and its interacting partners viz.; CCS, GPX1, GPX2, GPX3, GPX4, GPX5, GPX7, GPX8, CAT, PRDX1, TXN, SIRT3, FOXO3, and AKT1 were found to be involved in MAPK, p53 and foxo signaling pathways. Molecular pathways involved in association with SODs indicate reactive oxygen species (ROS) detoxification, apoptotic pathways and cell cycle regulation. Overall data revealed alleviated levels of SOD2 gene and decreased expression of SOD1 gene in response to C. trachomatis-infection leading to production of oxidative stress and RSA.
Collapse
|
47
|
Wu J, Lin S, Huang P, Qiu L, Jiang Y, Zhang Y, Meng N, Meng M, Wang L, Deng W, Liu Z, Guo C, Lu J, Wang H, Kong S. Maternal anxiety affects embryo implantation via impairing adrenergic receptor signaling in decidual cells. Commun Biol 2022; 5:840. [PMID: 35982177 PMCID: PMC9388523 DOI: 10.1038/s42003-022-03694-1] [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: 10/23/2021] [Accepted: 07/10/2022] [Indexed: 11/09/2022] Open
Abstract
Recurrent implantation failure (RIF) is defined as the failed pregnancy after good embryo transfer over 3 cycles during in vitro fertilization (IVF).The human endometrium plays a vital role in providing the site for embryo implantation, with several factors implicated in unsatisfactory endometrial receptivity in RIF. Our present results revealed that women with pregnancy loss or infertility have a higher serum epinephrine level, indicating a potential correlation between psychological stress and pregnancy failure. RNA-sequencing of the tissues collected at the endometrial receptive phase in normal and RIF women showed that stress hormones could affect the functional status of endometrial receptivity. Subsequent analysis revealed that the epinephrine signaling acts as an important regulator of endometrial receptivity through the PI3K-AKT and FOXO1 signaling pathways. We also found that patients with RIF show attenuated expression of the alpha-2C-adrenergic receptor (ADRA2C) and that its down regulation induced by high level epinephrine could inhibit the decidualization. Early pregnant mice treated with stress showed high serum epinephrine levels, defective uterine adrenergic receptor expression, and low pregnancy rates. Altogether, our findings indicate that mental stress during early pregnancy can alter the functional status of endometrial receptivity.
Collapse
Affiliation(s)
- Jinxiang Wu
- Department of Reproductive Medicine, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China.
| | - Shu Lin
- Centre of Neurological and Metabolic Research, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China.,Diabetes and Metabolism Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Pinxiu Huang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Lingling Qiu
- Department of Reproductive Medicine, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Yufei Jiang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Ying Zhang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Nan Meng
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Meiqing Meng
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Lemeng Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Wenbo Deng
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zhao Liu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Chuanhui Guo
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Jinhua Lu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Haibin Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.
| | - Shuangbo Kong
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
48
|
Cummings MJ, Yu H, Paudel S, Hu G, Li X, Hemberger M, Wang X. Uterine-specific SIRT1 deficiency confers premature uterine aging and impairs invasion and spacing of blastocyst, and stromal cell decidualization, in mice. Mol Hum Reprod 2022; 28:gaac016. [PMID: 35536234 PMCID: PMC10689003 DOI: 10.1093/molehr/gaac016] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/28/2022] [Indexed: 12/02/2023] Open
Abstract
A distinct age-related alteration in the uterine environment has recently been identified as a prevalent cause of the reproductive decline in older female mice. However, the molecular mechanisms that underlie age-associated uterine adaptability to pregnancy are not known. Sirtuin 1 (SIRT1), a multifunctional NAD+-dependent deacetylase that regulates cell viability, senescence and inflammation during aging, is reduced in aged decidua. Thus, we hypothesize that SIRT1 plays a critical role in uterine adaptability to pregnancy and that uterine-specific ablation of Sirt1 gene accelerates premature uterine aging. Female mice with uterine ablation of Sirt1 gene using progesterone receptor Cre (PgrCre) exhibit subfertility and signs of premature uterine aging. These Sirt1-deficient mothers showed decreases in litter size from their 1st pregnancy and became sterile (25.1 ± 2.5 weeks of age) after giving birth to the third litter. We report that uterine-specific Sirt1 deficiency impairs invasion and spacing of blastocysts, and stromal cell decidualization, leading to abnormal placentation. We found that these problems traced back to the very early stages of hormonal priming of the uterus. During the window of receptivity, Sirt1 deficiency compromises uterine epithelial-stromal crosstalk, whereby estrogen, progesterone and Indian hedgehog signaling pathways are dysregulated, hampering stromal cell priming for decidualization. Uterine transcriptomic analyses also link these causes to perturbations of histone proteins and epigenetic modifiers, as well as adrenomedullin signaling, hyaluronic acid metabolism, and cell senescence. Strikingly, our results also identified genes with significant overlaps with the transcriptome of uteri from aged mice and transcriptomes related to master regulators of decidualization (e.g. Foxo1, Wnt4, Sox17, Bmp2, Egfr and Nr2f2). Our results also implicate accelerated deposition of aging-related fibrillar Type I and III collagens in Sirt1-deficient uteri. Collectively, SIRT1 is an important age-related regulator of invasion and spacing of blastocysts, as well as decidualization of stromal cells.
Collapse
Affiliation(s)
- Magdalina J Cummings
- Department of Animal Science, North Carolina State University, Raleigh, NC, USA
- The Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Hongyao Yu
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Sudikshya Paudel
- Department of Animal Science, North Carolina State University, Raleigh, NC, USA
- The Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Guang Hu
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Xiaoling Li
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Myriam Hemberger
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Xiaoqiu Wang
- Department of Animal Science, North Carolina State University, Raleigh, NC, USA
- The Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
49
|
Hwang YJ, Sung GJ, Marquardt R, Young SL, Lessey BA, Kim TH, Cheon YP, Jeong JW. SIRT1 plays an important role in implantation and decidualization during mouse early pregnancy. Biol Reprod 2022; 106:1072-1082. [PMID: 35134122 PMCID: PMC9198957 DOI: 10.1093/biolre/ioac026] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 01/13/2022] [Accepted: 01/28/2022] [Indexed: 02/04/2023] Open
Abstract
Sirtuin 1 (SIRT1) is a member of the sirtuin family that functions to deacetylate both histones and non-histone proteins. Previous studies have identified significant SIRT1 upregulation in eutopic endometrium from infertile women with endometriosis. However, SIRT1 function in the uterus has not been directly studied. Using immunochemistry analysis, we found SIRT1 to be most strongly expressed at GD4.5 and GD5.5 in decidualized cells and at GD7.5 in secondary decidual cells in mouse. To assess the role of SIRT1 in uterine function, we generated uterine Sirt1 conditional knockout mice (Pgrcre/+Sirt1f/f; Sirt1d/d). A 6-month fertility trial revealed that Sirt1d/d females were subfertile. Implantation site numbers were significantly decreased in Sirt1d/d mice compared with controls at GD5.5. Sirt1d/d implantation sites at GD4.5 could be divided into two groups, Group #1 with luminal closure and nonspecific COX2 expression compared with controls (14/20) and Group #2 with an open lumen and no COX2 (6/20). In Sirt1d/d Group #1, nuclear FOXO1 expression in luminal epithelial cells was significantly decreased. In Sirt1d/d Group #2, nuclear FOXO1 expression was almost completely absent, and there was strong PGR expression in epithelial cells. At GD5.5, stromal PGR and COX2 were significantly decreased in Sirt1d/d uterine in the areas surrounding the embryo compared with controls, indicating defective decidualization. An artificially induced decidualization test revealed that Sirt1d/d females showed defects in decidualization response. All together, these data suggest that SIRT1 is important for decidualization and contributes to preparing a receptive endometrium for successful implantation.
Collapse
Affiliation(s)
- Yeon Jeong Hwang
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, USA
- Division of Developmental Biology and Physiology, Department of Biotechnology, Institute of Basic Sciences, Sungshin Women’s University, Seoul, South Korea
| | - Gi-Jun Sung
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, USA
| | - Ryan Marquardt
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, USA
- Cell and Molecular Biology Program, College of Natural Science, Michigan State University, East Lansing, MI, USA
| | - Steven L Young
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of North Carolina, Chapel Hill, NC, USA
| | - Bruce A Lessey
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertilithy, Atrium Health, Wake Forest Baptist, Winston-Salem, NC, USA
| | - Tae Hoon Kim
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, USA
| | - Yong-Pil Cheon
- Division of Developmental Biology and Physiology, Department of Biotechnology, Institute of Basic Sciences, Sungshin Women’s University, Seoul, South Korea
| | - Jae-Wook Jeong
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, USA
| |
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW To succinctly review the basic mechanisms of implantation and luteal phase endometrial differentiation, the etiologies of impaired endometrial function and receptivity, and the current methods that exist to evaluate and treat impaired endometrial receptivity. RECENT FINDINGS Human embryo implantation requires bidirectional communication between blastocyst and a receptive endometrium. Etiologies of impaired endometrial receptivity are varied. Some of these include delayed endometrial maturation, structural abnormalities, inflammation, and progesterone resistance. Current methods to evaluate endometrial receptivity include ultrasonography, hysteroscopy, and endometrial biopsy. Treatments are limited, but include operative hysteroscopy, treatment of endometriosis, and personalized timing of embryo transfer. SUMMARY Although some mechanisms of impaired endometrial receptivity are well understood, treatment options remain limited. Future efforts should be directed towards developing interventions targeted towards the known mediators of impaired endometrial receptivity.
Collapse
|