1
|
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
|
2
|
Dai W, Guo R, Na X, Jiang S, Liang J, Guo C, Fang Y, Na Z, Li D. Hypoxia and the endometrium: An indispensable role for HIF-1α as therapeutic strategies. Redox Biol 2024; 73:103205. [PMID: 38815332 PMCID: PMC11167393 DOI: 10.1016/j.redox.2024.103205] [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: 04/09/2024] [Revised: 04/30/2024] [Accepted: 05/20/2024] [Indexed: 06/01/2024] Open
Abstract
Hypoxia-inducible factor 1 alpha (HIF-1α) is a major molecular mediator of the hypoxic response. In the endometrium, local hypoxic conditions induced by hormonal fluctuations and endometrial vascular remodeling contribute to the production of HIF-1α, which plays an indispensable role in a series of physiological activities, such as menstruation and metamorphosis. The sensitive regulation of HIF-1α maintains the cellular viability and regenerative capacity of the endometrium against cellular stresses induced by hypoxia and excess reactive oxygen species. In contrast, abnormal HIF-1α levels exacerbate the development of various endometrial pathologies. This knowledge opens important possibilities for the development of promising HIF-1α-centered strategies to ameliorate endometrial disease. Nonetheless, additional efforts are required to elucidate the regulatory network of endometrial HIF-1α and promote the applications of HIF-1α-centered strategies in the human endometrium. Here, we summarize the role of the HIF-1α-mediated pathway in endometrial physiology and pathology, highlight the latest HIF-1α-centered strategies for treating endometrial diseases, and improve endometrial receptivity.
Collapse
Affiliation(s)
- Wanlin Dai
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Renhao Guo
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xinni Na
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shuyi Jiang
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Junzhi Liang
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Cuishan Guo
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuanyuan Fang
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China; NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, China.
| | - Zhijing Na
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China; NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, China.
| | - Da Li
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China; NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang, China; Key Laboratory of Reproductive Dysfunction Diseases and Fertility Remodeling of Liaoning Province, Shenyang, China.
| |
Collapse
|
3
|
Ammar AY, Minisy FM, Shawki HH, Mansour M, Hemeda SA, El Nahas AF, Sherif AH, Oishi H. Exposure to a Low-Oxygen Environment Causes Implantation Failure and Transcriptomic Shifts in Mouse Uteruses and Ovaries. Biomedicines 2024; 12:1016. [PMID: 38790978 PMCID: PMC11118081 DOI: 10.3390/biomedicines12051016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
Hypoxia is a condition in which tissues of the body do not receive sufficient amounts of oxygen supply. Numerous studies have elucidated the intricate roles of hypoxia and its involvement in both physiological and pathological conditions. This study aimed to clarify the impact of a forced low-oxygen environment in early pregnancy by exposing mice to low-oxygen conditions for 24-72 h after fertilization. The treatment resulted in the complete failure of blastocyst implantation, accompanied by vascular hyperpermeability in the uterus. A transcriptome analysis of the uterus revealed remarkable alterations in gene expression between control normoxic- and hypoxic-treatment groups. These alterations were characterized by the differentially expressed genes categorized into the immune responses and iron coordination. Furthermore, exposure to a low-oxygen environment caused apoptosis in the corpus luteum within the ovary and a reduction in progesterone secretion. Consequently, diminished plasma progesterone levels were considered to contribute to implantation failure in combination with the activation of the hypoxic pathway in the uterus. Additionally, previous studies have demonstrated the impact of hypoxic reactions on blastocyst development and the pre-implantation process in the endometrium. Our findings suggest that the corpus luteum exhibits elevated susceptibility to hypoxia, thereby elucidating a critical aspect of its physiological response.
Collapse
Affiliation(s)
- Asmaa Y. Ammar
- Biotechnology Department, Animal Health Research Institute (AHRI), Agriculture Research Center (ARC), Kafrelsheikh 12619, Egypt;
- Genetics Laboratory, Department of Animal Husbandry and Animal Wealth Development, Faculty of Veterinary Medicine, Alexandria University, Alexandria 21544, Egypt; (S.A.H.); (A.F.E.N.)
- Department of Comparative and Experimental Medicine, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan;
| | - Fatma M. Minisy
- Department of Comparative and Experimental Medicine, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan;
| | - Hossam H. Shawki
- Department of Comparative and Experimental Medicine, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan;
| | | | - Shabaan A. Hemeda
- Genetics Laboratory, Department of Animal Husbandry and Animal Wealth Development, Faculty of Veterinary Medicine, Alexandria University, Alexandria 21544, Egypt; (S.A.H.); (A.F.E.N.)
| | - Abeer F. El Nahas
- Genetics Laboratory, Department of Animal Husbandry and Animal Wealth Development, Faculty of Veterinary Medicine, Alexandria University, Alexandria 21544, Egypt; (S.A.H.); (A.F.E.N.)
| | - Ahmed H. Sherif
- Fish Disease Department, Animal Health Research Institute (AHRI), Agriculture Research Center (ARC), Kafrelsheikh 12619, Egypt
| | - Hisashi Oishi
- Department of Comparative and Experimental Medicine, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan;
| |
Collapse
|
4
|
Gudenschwager-Basso EK, Frydman G, Weerakoon S, Andargachew H, Piltaver CM, Huckle WR. Morphological evaluation of the feline placenta correlates with gene expression of vascular growth factors and receptors†. Biol Reprod 2024; 110:569-582. [PMID: 38092011 DOI: 10.1093/biolre/ioad167] [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: 01/03/2023] [Revised: 09/06/2023] [Accepted: 11/30/2023] [Indexed: 03/16/2024] Open
Abstract
Placental angiogenesis is critical for normal development. Angiogenic factors and their receptors are key regulators of this process. Dysregulated placental vascular development is associated with pregnancy complications. Despite their importance, vascular growth factor expression has not been thoroughly correlated with placental morphologic development across gestation in cats. We postulate that changes in placental vessel morphology can be appreciated as consequences of dynamic expression of angiogenic signaling agents. Here, we characterized changes in placental morphology alongside expression analysis of angiogenic factor splice variants and receptors throughout pregnancy in domestic shorthair cats. We observed increased vascular and lamellar density in the lamellar zone during mid-pregnancy. Immunohistochemical analysis localized the vascular endothelial growth factor A (VEGF-A) receptor KDR to endothelial cells of the maternal and fetal microvasculatures. PlGF and its principal receptor Flt-1 were localized to the trophoblasts and fetal vasculature. VEGF-A was found in trophoblast cells and associated with endothelial cells. We detected expression of two Plgf splice variants and four Vegf-a variants. Quantitative real-time polymerase chain reaction analysis showed upregulation of mRNAs encoding pan Vegf-a and all Vegf-a splice forms at gestational days 30-35. Vegf-A showed a marked relative increase in expression during mid-pregnancy, consistent with the pro-angiogenic changes seen in the lamellar zone at days 30-35. Flt-1 was upregulated during late pregnancy. Plgf variants showed stable expression during the first two-thirds of pregnancy, followed by a marked increase toward term. These findings revealed specific spatiotemporal expression patterns of VEGF-A family members consistent with pivotal roles during normal placental development.
Collapse
Affiliation(s)
- Erwin K Gudenschwager-Basso
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, USA
| | - Galit Frydman
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, USA
| | - Shaneke Weerakoon
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, USA
- Virginia Tech Carilion School of Medicine, Virginia Tech, Blacksburg, VA, USA
| | - Hariyat Andargachew
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, USA
| | - Cassandra M Piltaver
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, USA
| | - William R Huckle
- Department of Biomedical Sciences & Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, USA
| |
Collapse
|
5
|
Turgut AO, Korkmaz Ağaoğlu Ö. Differential expression of angiogenesis-related genes in goat uterus and corpus luteum during pregnancy. Reprod Domest Anim 2023; 58:1672-1684. [PMID: 37776186 DOI: 10.1111/rda.14482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/14/2023] [Accepted: 09/19/2023] [Indexed: 10/01/2023]
Abstract
Vascularization and the control of luteal and endometrial development are regulated by hypoxia-inducible factors (HIFs) and vascular endothelial growth factor (VEGF) during pregnancy. In this study, the mRNA and protein expression levels of HIFs (HIF1A, HIF2A and HIF3A) and VEGF in goat uterine and ovarian tissues during various stages of pregnancy were evaluated. A total of 42 Hair goats were used and were allocated into six groups, namely embryo-positive (G1), early pregnancy (G2), mid-term pregnancy (G3), late pregnancy (G4), oocyte-positive group (G5) and diestrus group (G6). The mRNA expression of the examined genes was evaluated by RT-qPCR, and protein expression was evaluated by immunohistochemistry (IHC). In caruncles, HIF1A mRNA expression was greater in G1, G2 and G4 than in G3 (p < .05). HIF1A and HIF2A expression was greater in G1 than in G5 (p < .05). In cotyledons, HIF1A, HIF2A and HIF3A mRNA expression was greater in G2 and G3 compared to G4 (p < .05). In luteal tissue, HIF1A mRNA expression was greater in G1 and G2 than in G3 and G4 (p < .05). In the immunohistochemical examination, HIF1A, HIF2A, HIF3A and VEGF immunoreactions were detected in uterine and luteal tissues. Findings suggest that HIFs and VEGF are involved in the regulation of ovarian functions as well as the processes of implantation and placentation.
Collapse
Affiliation(s)
- Ali Osman Turgut
- Department of Animal Science, Faculty of Veterinary Medicine, Siirt University, Siirt, Turkey
| | - Özgecan Korkmaz Ağaoğlu
- Department of Genetics, Faculty of Veterinary Medicine, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| |
Collapse
|
6
|
Varrias D, Spanos M, Kokkinidis DG, Zoumpourlis P, Kalaitzopoulos DR. Venous Thromboembolism in Pregnancy: Challenges and Solutions. Vasc Health Risk Manag 2023; 19:469-484. [PMID: 37492280 PMCID: PMC10364824 DOI: 10.2147/vhrm.s404537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 07/13/2023] [Indexed: 07/27/2023] Open
Abstract
Venous thromboembolism (VTE) is a serious medical condition that can lead to severe morbidity and mortality, making it a significant public health concern. VTE is a multifactorial condition that results from the interaction of genetic, acquired, and environmental factors. Physiological changes during pregnancy increase the risk of VTE as they express Virchow's triad (increased coagulation factors, decreased fibrinolysis, trauma, and venous stasis). Moreover, pregnancy-related risk factors, such as advanced maternal age, obesity, multiple gestations, and cesarean delivery, further increase the risk of VTE. Managing VTE in pregnancy is challenging due to the complexity of balancing the risks and benefits of anticoagulant therapy for both the mother and the fetus. A multidisciplinary approach involving obstetricians, hematologists, and neonatologists, is necessary to ensure optimal outcomes for both the mother and baby. This review aims to discuss the current challenges associated with VTE in pregnancy and identify potential solutions for improving outcomes for pregnant women at risk for VTE.
Collapse
Affiliation(s)
- Dimitrios Varrias
- Department of Medicine, Jacobi Medical Center, Bronx, NY, 10461, USA
- Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Michail Spanos
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Damianos G Kokkinidis
- Section of Cardiovascular Medicine, Yale University School of Medicine, Yale New Haven Hospital, New Haven, CT, USA
| | - Panagiotis Zoumpourlis
- Department of Medicine, Jacobi Medical Center, Bronx, NY, 10461, USA
- Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | | |
Collapse
|
7
|
Seki M, Takeuchi E, Fukui E, Matsumoto H. Upregulation of iNOS and phosphorylated eNOS in the implantation-induced blastocysts of mice. Reprod Med Biol 2023; 22:e12545. [PMID: 37841392 PMCID: PMC10568119 DOI: 10.1002/rmb2.12545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/20/2023] [Accepted: 09/16/2023] [Indexed: 10/17/2023] Open
Abstract
Purpose This study aimed to examine expressions of iNOS and phosphorylated eNOS (p-eNOS) in implantation-induced blastocysts. We also examined the upstream of p-eNOS. Methods To address the protein expressions in implantation-induced blastocysts, we performed immunohistochemical analysis using a delayed implantation mouse model. Immunostaining for iNOS, p-eNOS, and p-Akt was done. To address the relationship between p-eNOS and p-Akt, activated blastocysts were treated with an Akt inhibitor, MK-2206. Results iNOS expression was at low levels in dormant blastocysts, whereas the expression was significantly increased in the activated blastocysts. Double staining of p-eNOS and p-Akt in individual blastocysts showed colocalization of p-eNOS and p-Akt of the trophectoderm. p-eNOS and p-Akt expressions were at low levels in dormant blastocysts, whereas both of them were significantly increased in the activated blastocysts. Both dormant and activated blastocysts showed significant positive correlations between p-eNOS and p-Akt. MK-2206 treatment for activated blastocysts showed that blastocysts with lower p-Akt had significantly lower p-eNOS levels. Conclusions iNOS and p-eNOS, Ca2+ independent NOS, are upregulated by E2 in the blastocysts during implantation activation. Furthermore, p-eNOS is upregulated in implantation-induced blastocysts downstream of p-Akt.
Collapse
Affiliation(s)
- Misato Seki
- Laboratory of Animal Breeding and Reproduction, Division of Animal Science, School of AgricultureUtsunomiya UniversityUtsunomiya, TochigiJapan
| | - Eisaku Takeuchi
- Laboratory of Animal Breeding and Reproduction, Division of Animal Science, School of AgricultureUtsunomiya UniversityUtsunomiya, TochigiJapan
| | - Emiko Fukui
- Laboratory of Animal Breeding and Reproduction, Division of Animal Science, School of AgricultureUtsunomiya UniversityUtsunomiya, TochigiJapan
- Center for Bioscience Research and EducationUtsunomiya UniversityUtsunomiya, TochigiJapan
| | - Hiromichi Matsumoto
- Laboratory of Animal Breeding and Reproduction, Division of Animal Science, School of AgricultureUtsunomiya UniversityUtsunomiya, TochigiJapan
- Center for Bioscience Research and EducationUtsunomiya UniversityUtsunomiya, TochigiJapan
| |
Collapse
|
8
|
Ma Q, Beal JR, Song X, Bhurke A, Bagchi IC, Bagchi MK. Extracellular Vesicles Secreted by Mouse Decidual Cells Carry Critical Information for the Establishment of Pregnancy. Endocrinology 2022; 163:6758297. [PMID: 36219207 PMCID: PMC9761388 DOI: 10.1210/endocr/bqac165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Indexed: 11/19/2022]
Abstract
The mouse decidua secretes many factors that act in a paracrine/autocrine manner to critically control uterine decidualization, neovascularization, and tissue remodeling that ensure proper establishment of pregnancy. The precise mechanisms that dictate intercellular communications among the uterine cells during early pregnancy remain unknown. We recently reported that conditional deletion of the gene encoding the hypoxia-inducible transcription factor 2 alpha (Hif2α) in mouse uterus led to infertility. Here, we report that HIF2α in mouse endometrial stromal cells (MESCs) acts via the cellular trafficking regulator RAB27b to control the secretion of extracellular vesicles (EVs) during decidualization. We also found that Hif2α-regulated pathways influence the biogenesis of EVs. Proteomic analysis of EVs secreted by decidualizing MESCs revealed that they harbor a wide variety of protein cargoes whose composition changed as the decidualization process progressed. The EVs enhanced the differentiation capacity of MESCs and the production of angiogenic factors by these cells. We also established that matrix metalloproteinase-2, a prominent EV cargo protein, modulates uterine remodeling during decidualization. Collectively, our results support the concept that EVs are central to the mechanisms by which the decidual cells communicate with each other and other cell types within the uterus to facilitate successful establishment of pregnancy.
Collapse
Affiliation(s)
- Qiuyan Ma
- Department of Molecular and Integrative Physiology, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA
| | - Jacob R Beal
- Department of Molecular and Integrative Physiology, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA
| | - Xiangning Song
- Department of Molecular and Integrative Physiology, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA
| | - Arpita Bhurke
- Carle Woese Institute of Genomic Biology, University of Illinois, Urbana-Champaign, Urbana, IL 61801, USA
| | - Indrani C Bagchi
- Correspondence: Indrani C. Bagchi, PhD, Departments of Comparative Biosciences, University of Illinois, Urbana-Champaign, Urbana, IL, USA. ; or Milan K. Bagchi, PhD, Departments of Molecular and Integrative Physiology, University of Illinois, Urbana-Champaign, Urbana, IL, USA.
| | - Milan K Bagchi
- Correspondence: Indrani C. Bagchi, PhD, Departments of Comparative Biosciences, University of Illinois, Urbana-Champaign, Urbana, IL, USA. ; or Milan K. Bagchi, PhD, Departments of Molecular and Integrative Physiology, University of Illinois, Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
9
|
Wang X, Miao S, Lu L, Yuan J, Pan S, Wu X. miR‑519d‑3p released by human blastocysts negatively regulates endometrial epithelial cell adhesion by targeting HIF1α. Int J Mol Med 2022; 50:123. [PMID: 35959792 PMCID: PMC9387561 DOI: 10.3892/ijmm.2022.5179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 07/04/2022] [Indexed: 11/06/2022] Open
Abstract
Successful embryo implantation requires a competent embryo, a receptive endometrium and synchronized communication between them. The selection of embryos with the highest implantation potential remains a challenge in the field of assisted reproductive technology. Moreover, little is known about the precise molecular mechanisms underlying embryo‑endometrium crosstalk. MicroRNAs (miRNAs/miRs) have been detected in the spent embryo culture medium (SCM); however, their functions at the preimplantation stage remain unclear. In the present study, human SCM samples were collected during in vitro fertilization/intracytoplasmic sperm injection‑embryo transfer and divided into implanted and not‑implanted groups according to the clinical pregnancy outcomes. Total RNA was extracted and six miRNAs (miR‑372‑3p, miR‑373‑3p, miR‑516b‑5p, miR‑517a‑3p, miR‑519d‑3p and miR‑520a‑3p) were selected for reverse transcription‑quantitative PCR (RT‑qPCR) analysis. The results revealed that miR‑372‑3p and miR‑519d‑3p were markedly increased in SCM from blastocysts that failed to implant compared with in blastocysts that implanted. The receiver operating characteristic curve analysis revealed that miR‑519d‑3p was superior to miR‑372‑3p in predicting pregnancy outcomes. In vitro miRNA uptake and cell adhesion assays were performed to determine whether miR‑519d‑3p could be taken up by endometrial epithelial cells and to examine the biological roles of miR‑519d‑3p after internalization. Potential targets of miR‑519d‑3p were verified using a dual‑luciferase reporter system. The results demonstrated that miR‑519d‑3p was taken up by human endometrial epithelial cells and that it may inhibit embryo adhesion by targeting HIF1α. Using RT‑qPCR, western blot analysis and flow cytometry assay, HIF1α was shown to inhibit the biosynthesis of fucosyltransferase 7 and sialyl‑Lewis X (sLex), a cell‑surface oligosaccharide that serves an important role in embryonic apposition and adhesion. In addition, a mouse model was established and the results suggested that miR‑519d‑3p overexpression hampered embryo implantation in vivo. Taken together, miRNAs in SCM may serve as novel biomarkers for embryo quality. Furthermore, miR‑519d‑3p was shown to mediate embryo‑endometrium crosstalk and to negatively regulate embryo implantation by targeting HIF1α/FUT7/sLex pathway.
Collapse
Affiliation(s)
- Xiaodan Wang
- Department of Obstetrics and Gynecology, Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Suibing Miao
- Reproductive Medicine Center, The Fourth Hospital of Shijiazhuang Affiliated to Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Linqi Lu
- Reproductive Medicine Center, The Fourth Hospital of Shijiazhuang Affiliated to Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Jingchuan Yuan
- Reproductive Medicine Center, The Fourth Hospital of Shijiazhuang Affiliated to Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Shuhong Pan
- Reproductive Medicine Center, The Fourth Hospital of Shijiazhuang Affiliated to Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Xiaohua Wu
- Department of Obstetrics and Gynecology, Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| |
Collapse
|
10
|
The Regulators of Human Endometrial Stromal Cell Decidualization. Biomolecules 2022; 12:biom12091275. [PMID: 36139114 PMCID: PMC9496326 DOI: 10.3390/biom12091275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/04/2022] [Accepted: 09/07/2022] [Indexed: 11/17/2022] Open
Abstract
Several factors are important for implantation and subsequent placentation in the endometrium, including immunity, angiogenesis, extracellular matrix, glucose metabolism, reactive oxidative stress, and hormones. The involvement or abnormality of these factors can impair canonical decidualization. Unusual decidualization can lead to perinatal complications, such as disruption of trophoblast invasion. Drastic changes in the morphology and function of human endometrial stromal cells (hESCs) are important for decidualization of the human endometrium; hESCs are used to induce optimal morphological and functional decidualization in vitro because they contain estrogen and progesterone receptors. In this review, we will focus on the studies that have been conducted on hESC decidualization, including the results from our laboratory.
Collapse
|
11
|
Zhang T, Shen HH, Qin XY, Li MQ. The metabolic characteristic of decidual immune cells and their unique properties in pregnancy loss. Immunol Rev 2022; 308:168-186. [PMID: 35582842 DOI: 10.1111/imr.13085] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/03/2022] [Indexed: 12/14/2022]
Abstract
Maternal tolerance to semi- or fully allograft conceptus is a prerequisite for the maintenance of pregnancy. Once this homeostasis is disrupted, it may result in pregnancy loss. As a potential approach to prevent pregnancy loss, targeting decidual immune cells (DICs) at the maternal-fetal interface has been suggested. Although the phenotypic features and functions of DIC have been extensively profiled, the regulatory pathways for this unique immunological adaption have yet to be elucidated. In recent years, a pivotal mechanism has been highlighted in the area of immunometabolism, by which the changes in intracellular metabolic pathways in DIC and interaction with the adjacent metabolites in the microenvironment can alter their phenotypes and function. More inspiringly, the manipulation of metabolic profiling in DIC provides a novel avenue for the prevention and treatment of pregnancy loss. Herein, this review highlights the major metabolic programs (specifically, glycolysis, ATP-adenosine metabolism, lysophosphatidic acid metabolism, and amino acid metabolism) in multiple immune cells (including decidual NK cells, macrophages, and T cells) and their integrations with the metabolic microenvironment in normal pregnancy. Importantly, this perspective may help to provide a potential therapeutic strategy for reducing pregnancy loss via targeting this interplay.
Collapse
Affiliation(s)
- Tao Zhang
- Assisted Reproductive Technology Unit, Department of Obstetrics and Gynecology, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
| | - Hui-Hui Shen
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China
| | - Xue-Yun Qin
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China
| | - Ming-Qing Li
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, China.,NHC Key Lab of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research, Fudan University, Shanghai, China.,Shanghai Medical School, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| |
Collapse
|
12
|
Zheng X, Ma J, Hu M, Long J, Wei Q, Ren W. Analysis of HIF2α polymorphisms in infertile women with polycystic ovary syndrome or unexplained infertility. Front Endocrinol (Lausanne) 2022; 13:986567. [PMID: 36157441 PMCID: PMC9492870 DOI: 10.3389/fendo.2022.986567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/17/2022] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVE To evaluate HIF2α polymorphisms and glucose metabolism in a group of women with polycystic ovary syndrome (PCOS) or unexplained infertility (UI). PATIENTS The infertile group (n=148) consisted of 96 women with PCOS, 52 women with UI, and176 women without infertility as a healthy control group. INTERVENTION We genotyped 29 single nucleotide polymorphisms (SNPs) of HIF2α by using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS)-based genotyping technology. The genetic associations were analyzed statistically. MAIN OUTCOME MEASURES Allele frequency, genotype distribution and haplotype analyze of the HIF2α polymorphisms were performed. Body mass index (BMI), waist circumference, uric acid (UA), high-sensitivity C-reactive protein (hsCRP), lipids, glucose and insulin tolerance - were also measured. RESULTS Infertile women with PCOS had a higherBMI and waist circumference, elevated hsCRP and uric acid (UA) levels, impaired glucose tolerance, and increased levels of plasma insulin compared to UI patients and healthy women. SNP analysis of HIF2α revealed that the allele and genotype frequencies of rs4953361 were significantly associated with infertile women with PCOS. Haplotype analysis of the HIF2α polymorphism identified haplotypes TGG and TGA as being associated with infertile women with PCOS. Women with the AA genotype of rs4953361 had a significantly higher BMI and post load plasma glucose and insulin levels than those of women with the GG genotype. CONCLUSION Infertile women with PCOS more commonly have metabolic disturbances than those with UI. This is the first study to report an association between HIF2α polymorphisms (rs4953361) and the risk of infertile women with PCOS, not UI, in Han Chinese population. These results require replication in larger populations.In this observational study, we did not report the results of a health care intervention on human participants. The study was approved by the Human Research Ethics Committee of the First Affiliated Hospital of Chongqing Medical University. Clinical data and peripheral blood samples were collected only after explaining the objectives of the study and obtaining a signed informed consent form.
Collapse
Affiliation(s)
- Xiaoya Zheng
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- *Correspondence: Xiaoya Zheng,
| | - Jiani Ma
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Min Hu
- Reproductive and Infertility Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jian Long
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qiang Wei
- Prevention of Disease Department, Chongqing Jiulongpo District Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Wei Ren
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
13
|
Maclean A, Adishesh M, Button L, Richards L, Alnafakh R, Newton E, Drury J, Hapangama DK. The effect of pre-analytical variables on downstream application and data analysis of human endometrial biopsies. Hum Reprod Open 2022; 2022:hoac026. [PMID: 35775066 PMCID: PMC9240853 DOI: 10.1093/hropen/hoac026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 05/27/2022] [Indexed: 11/14/2022] Open
Abstract
STUDY QUESTION What are the effects of pre-analytical variables on the downstream analysis of patient-derived endometrial biopsies? SUMMARY ANSWER There are distinct differences in the protein levels of the master regulator of oxygen homeostasis, hypoxia-inducible factor-1-alpha (HIF1α), and the protein and mRNA levels of three related genes, carbonic anhydrase 9 (CA9), vascular endothelial growth factor A (VEGFA) and progesterone receptor (PR) in human endometrial biopsies, depending on the pre-analytical variables: disease status (cancer vs benign), timing of biopsy (pre- vs post-hysterectomy) and type of biopsy (pipelle vs full-thickness). WHAT IS KNOWN ALREADY Patient-derived biopsies are vital to endometrial research, but pre-analytical variables relating to their collection may affect downstream analysis, as is evident in other tissues. STUDY DESIGN SIZE DURATION A prospective observational study including patients undergoing hysterectomy for endometrial cancer (EC) or benign indications was conducted at a large tertiary gynaecological unit in the UK. Endometrial biopsies were obtained at different time points (pre- or post-hysterectomy) using either a pipelle endometrial sampler or as a full-thickness wedge biopsy. PARTICIPANTS/MATERIALS SETTING METHODS The changes in HIF1α, CA9, VEGFA and PR protein levels were measured by semi-quantitative analysis of immunostaining, and the expression levels of three genes (CA9, VEGFA and PR) were investigated by quantitative real-time PCR, in endometrial biopsies from 43 patients undergoing hysterectomy for EC (n = 22) or benign gynaecological indications (n = 21). MAIN RESULTS AND THE ROLE OF CHANCE An increase in HIF1α immunostaining was observed in EC versus benign endometrium (functionalis glands) obtained pre-hysterectomy (P < 0.001). An increase in CA9 immunostaining was observed in EC versus benign endometrial functionalis glands at both pre- and post-hysterectomy time points (P = 0.03 and P = 0.003, respectively). Compared with benign endometrial pipelle samples, EC samples demonstrated increased mRNA expression of CA9 (pre-hysterectomy P < 0.001, post-hysterectomy P = 0.008) and VEGFA (pre-hysterectomy P = 0.004, post-hysterectomy P = 0.002). In benign uteri, HIF1α immunoscores (functionalis glands, P = 0.03 and stroma, P = 0.009), VEGFA immunoscores (functionalis glands, P = 0.03 and stroma, P = 0.01) and VEGFA mRNA levels (P = 0.008) were increased in matched post-hysterectomy versus pre-hysterectomy samples. Similarly, in EC, an increase in VEGFA immunoscores (epithelial and stromal) and VEGFA mRNA expression was observed in the matched post-hysterectomy versus pre-hysterectomy biopsies (P = 0.008, P = 0.004 and P = 0.018, respectively). Full-thickness benign post-hysterectomy endometrial biopsies displayed increased VEGFA (P = 0.011) and PR (P = 0.006) mRNA expression compared with time-matched pipelle biopsies. LARGE SCALE DATA N/A. LIMITATIONS REASONS FOR CAUTION This descriptive study explores the effect of pre-analytical variables on the expression of four proteins and three hypoxia-related genes in a limited number of endometrial biopsies from patients with EC and benign controls. Due to the small number, it was not possible to investigate other potential variables such as menstrual cycle phase, region-specific differences within the endometrium, grade and stage of cancer, and surgical technicalities. WIDER IMPLICATIONS OF THE FINDINGS Careful consideration of the effects of these pre-analytical variables is essential when interpreting data relating to human endometrial biopsies. A standardized approach to endometrial tissue collection is essential to ensure accurate and clinically transferrable data. STUDY FUNDING/COMPETING INTERESTS The authors have no conflicts of interest to declare. The work included in this manuscript was funded by Wellbeing of Women project grants RG1073 and RG2137 (D.K.H.), Wellbeing of Women Entry-Level Scholarship ELS706 and Medical Research Council MR/V007238/1 (A.M./D.K.H.), Liverpool Women's Hospital Cancer Charity (M.A.) and University of Liverpool (L.B., L.R. and E.N.).
Collapse
Affiliation(s)
- A Maclean
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - M Adishesh
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - L Button
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - L Richards
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - R Alnafakh
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - E Newton
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - J Drury
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - D K Hapangama
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| |
Collapse
|
14
|
Bishop A, Cartwright JE, Whitley GS. Stanniocalcin-1 in the female reproductive system and pregnancy. Hum Reprod Update 2021; 27:1098-1114. [PMID: 34432025 PMCID: PMC8542996 DOI: 10.1093/humupd/dmab028] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 06/15/2021] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Stanniocalcin-1 (STC-1) is a widely expressed glycoprotein hormone involved in a diverse spectrum of physiological and pathophysiological processes including angiogenesis, mineral homeostasis, cell proliferation, inflammation and apoptosis. Over the last 20 years, numerous studies have reported STC-1 expression within female reproductive tissues including the uterus, ovaries and placenta and implicated STC-1 in processes such as ovarian follicular development, blastocyst implantation, vascular remodelling in early pregnancy and placental development. Notably, dysregulation of STC-1 within reproductive tissues has been linked to the onset of severe reproductive disorders including endometriosis, polycystic ovary syndrome, poor trophoblast invasion and placental perfusion in early pregnancy. Furthermore, significant changes in tissue expression and in maternal systemic concentration take place throughout pregnancy and further substantiate the vital role of this protein in reproductive health and disease. OBJECTIVE AND RATIONALE Our aim is to provide a comprehensive overview of the existing literature, to summarise the expression profile and roles of STC-1 within the female reproductive system and its associated pathologies. We highlight the gaps in the current knowledge and suggest potential avenues for future research. SEARCH METHODS Relevant studies were identified through searching the PubMed database using the following search terms: ‘stanniocalcin-1’, ‘placenta’, ‘ovary’, ‘endometrium’, ‘pregnancy’, ‘reproduction’, ‘early gestation’. Only English language papers published between 1995 and 2020 were included. OUTCOMES This review provides compelling evidence of the vital function that STC-1 plays within the female reproductive system. The literature presented summarise the wide expression profile of STC-1 within female reproductive organs, as well as highlighting the putative roles of STC-1 in various functions in the reproductive system. Moreover, the observed link between altered STC-1 expression and the onset of various reproductive pathologies is presented, including those in pregnancy whose aetiology occurs in the first trimester. This summary emphasises the requirement for further studies on the mechanisms underlying the regulation of STC-1 expression and function. WIDER IMPLICATIONS STC-1 is a pleiotropic hormone involved in the regulation of a number of important biological functions needed to maintain female reproductive health. There is also growing evidence that dysregulation of STC-1 is implicated in common reproductive and obstetric disorders. Greater understanding of the physiology and biochemistry of STC-1 within the field may therefore identify possible targets for therapeutic intervention and/or diagnosis.
Collapse
Affiliation(s)
- Alexa Bishop
- Centre for Vascular Biology, Molecular and Clinical Sciences Research Institute, St George's, University of London, London, UK
| | - Judith E Cartwright
- Centre for Vascular Biology, Molecular and Clinical Sciences Research Institute, St George's, University of London, London, UK
| | - Guy S Whitley
- Centre for Vascular Biology, Molecular and Clinical Sciences Research Institute, St George's, University of London, London, UK
| |
Collapse
|
15
|
Defective Uteroplacental Vascular Remodeling in Preeclampsia: Key Molecular Factors Leading to Long Term Cardiovascular Disease. Int J Mol Sci 2021; 22:ijms222011202. [PMID: 34681861 PMCID: PMC8539609 DOI: 10.3390/ijms222011202] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/15/2021] [Accepted: 10/16/2021] [Indexed: 12/12/2022] Open
Abstract
Preeclampsia is a complex hypertensive disorder in pregnancy which can be lethal and is responsible for more than 70,000 maternal deaths worldwide every year. Besides the higher risk of unfavorable obstetric outcomes in women with preeclampsia, another crucial aspect that needs to be considered is the association between preeclampsia and the postpartum cardiovascular health of the mother. Currently, preeclampsia is classified as one of the major risk factors of cardiovascular disease (CVD) in women, which doubles the risk of venous thromboembolic events, stroke, and ischemic heart disease. In order to comprehend the pathophysiology behind the linkage between preeclampsia and the development of postpartum CVD, a thorough understanding of the abnormal uteroplacental vascular remodeling in preeclampsia is essential. Therefore, this review aims to summarize the current knowledge of the defective process of spiral artery remodeling in preeclampsia and how the resulting placental damage leads to excessive angiogenic imbalance and systemic inflammation in long term CVD. Key molecular factors in the pathway-including novel findings of microRNAs-will be discussed with suggestions of future management strategies of preventing CVD in women with a history of preeclampsia.
Collapse
|
16
|
Biology and pathology of the uterine microenvironment and its natural killer cells. Cell Mol Immunol 2021; 18:2101-2113. [PMID: 34426671 PMCID: PMC8429689 DOI: 10.1038/s41423-021-00739-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/12/2021] [Indexed: 02/07/2023] Open
Abstract
Tissues are the new frontier of discoveries in immunology. Cells of the immune system are an integral part of tissue physiology and immunity. Determining how immune cells inhabit, housekeep, and defend gut, lung, brain, liver, uterus, and other organs helps revealing the intimate details of tissue physiology and may offer new therapeutic targets to treat pathologies. The uterine microenvironment modulates the development and function of innate lymphoid cells [ILC, largely represented by natural killer (NK) cells], macrophages, T cells, and dendritic cells. These immune cells, in turn, contribute to tissue homeostasis. Regulated by ovarian hormones, the human uterine mucosa (endometrium) undergoes ~400 monthly cycles of breakdown and regeneration from menarche to menopause, with its fibroblasts, glands, blood vessels, and immune cells remodeling the tissue into the transient decidua. Even more transformative changes occur upon blastocyst implantation. Before the placenta is formed, the endometrial glands feed the embryo by histiotrophic nutrition while the uterine spiral arteries are stripped of their endothelial layer and smooth muscle actin. This arterial remodeling is carried out by invading fetal trophoblast and maternal immune cells, chiefly uterine NK (uNK) cells, which also assist fetal growth. The transformed arteries no longer respond to maternal stimuli and meet the increasing demands of the growing fetus. This review focuses on how the everchanging uterine microenvironment affects uNK cells and how uNK cells regulate homeostasis of the decidua, placenta development, and fetal growth. Determining these pathways will help understand the causes of major pregnancy complications.
Collapse
|
17
|
Lee B, Shin H, Oh JE, Park J, Park M, Yang SC, Jun JH, Hong SH, Song H, Lim HJ. An autophagic deficit in the uterine vessel microenvironment provokes hyperpermeability through deregulated VEGFA, NOS1, and CTNNB1. Autophagy 2021; 17:1649-1666. [PMID: 32579471 PMCID: PMC8354601 DOI: 10.1080/15548627.2020.1778292] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/22/2020] [Accepted: 05/02/2020] [Indexed: 02/07/2023] Open
Abstract
The uterus undergoes vascular changes during the reproductive cycle and pregnancy. Steroid hormone deprivation induces macroautophagy/autophagy in major uterine cell types. Herein, we explored the functions of uterine autophagy using the Amhr2-Cre-driven atg7 deletion model. Deletion of Atg7 was confirmed by functional deficit of autophagy in uterine stromal, myometrial, and vascular smooth muscle cells, but not in endothelial cells. atg7d/d uteri exhibited enhanced stromal edema accompanied by dilation of blood vessels. Ovariectomized atg7d/d uteri showed decreased expression of endothelial junction-related proteins, such as CTNNB1/beta-catenin, with increased vascular permeability, and increased expression of VEGFA and NOS1. Nitric oxide (NO) was shown to mediate VEGFA-induced vascular permeability by targeting CTNNB1. NO involvement in maintaining endothelial junctional stability in atg7d/d uteri was confirmed by the reduction in extravasation following treatment with a NOS inhibitor. We also showed that atg7d/d uterine phenotype improved the fetal weight:placental weight ratio, which is one of the indicators of assessing the status of preeclampsia. We showed that autophagic deficit in the uterine vessel microenvironment provokes hyperpermeability through the deregulation of VEGFA, NOS1, and CTNNB1.Abbreviations: ACTA2: actin, alpha 2, smooth muscle, aortic; Amhr2: anti-Mullerian hormone type 2 receptor; ANGPT1: angiopoietin 1; ATG: autophagy-related; CDH5: cadherin 5; CLDN5: claudin 5; COL1A1: collagen, type I, alpha 1; CSPG4/NG2: chondroitin sulfate proteoglycan 4; CTNNB1: catenin (cadherin associated protein), beta 1; DES: desmin; EDN1: endothelin 1; EDNRB: endothelin receptor type B; F3: coagulation factor III; KDR/FLK1/VEGFR2: kinase insert domain protein receptor; LYVE1: lymphatic vessel endothelial hyaluronan receptor 1; MAP1LC3B: microtubule-associated protein 1 light chain 3 beta; MCAM/CD146: melanoma cell adhesion molecule; MYL2: myosin, light polypeptide 2, regulatory, cardiac, slow; MYLK: myosin, light polypeptide kinase; NOS1/nNOS: nitric oxide synthase 1, neuronal; NOS2/iNOS: nitric oxide synthase 2, inducible; NOS3/eNOS: nitric oxide synthase 3, endothelial cell; OVX: ovariectomy; PECAM1/CD31: platelet/endothelial cell adhesion molecule 1; POSTN: periostin, osteoblast specific factor; SQSTM1: sequestosome 1; TEK/Tie2: TEK receptor tyrosine kinase; TJP1/ZO-1: tight junction protein 1; TUBB1, tubulin, beta 1 class VI; USC: uterine stromal cell; VEGFA: vascular endothelial growth factor A; VSMC: vascular smooth muscle cell.
Collapse
Affiliation(s)
- Bora Lee
- Department of Biomedical Science & Technology, Konkuk University, Seoul, Korea
| | - Hyejin Shin
- Department of Biomedical Science & Technology, Konkuk University, Seoul, Korea
| | - Ji-Eun Oh
- Department of Veterinary Medicine, Konkuk University, Seoul, Korea
| | - Jaekyoung Park
- Department of Biomedical Science & Technology, Konkuk University, Seoul, Korea
| | - Mira Park
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi-do, Korea
| | - Seung Chel Yang
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi-do, Korea
| | - Jin-Hyun Jun
- Department of Biomedical Laboratory Science, Eulji University, Seongnam, Gyeonggi-do, Korea
- Department of Senior Healthcare, BK21 Plus Program, Eulji Medi-Bio Research Institute, Graduate School, Eulji University, Daejeon, Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Kangwon-do, Chuncheon, Korea
| | - Haengseok Song
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi-do, Korea
| | - Hyunjung Jade Lim
- Department of Biomedical Science & Technology, Konkuk University, Seoul, Korea
- Department of Veterinary Medicine, Konkuk University, Seoul, Korea
| |
Collapse
|
18
|
Khatun M, Arffman RK, Lavogina D, Kangasniemi M, Laru J, Ahtikoski A, Lehtonen S, Paulson M, Hirschberg AL, Salumets A, Andersson LC, Piltonen TT. Women with polycystic ovary syndrome present with altered endometrial expression of stanniocalcin-1†. Biol Reprod 2021; 102:306-315. [PMID: 31494675 PMCID: PMC7016287 DOI: 10.1093/biolre/ioz180] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 08/06/2019] [Accepted: 08/30/2019] [Indexed: 12/13/2022] Open
Abstract
Stanniocalcin-1 (STC-1) is a pro-survival factor that protects tissues against stressors, such as hypoxia and inflammation. STC-1 is co-expressed with the endometrial receptivity markers, and recently endometrial STC-1 was reported to be dysregulated in endometriosis, a condition linked with endometrial progesterone resistance and inflammation. These features are also common in the endometrium in women with polycystic ovary syndrome (PCOS), the most common endocrine disorder in women. Given that women with PCOS present with subfertility, pregnancy complications, and increased risk for endometrial cancer, we investigated endometrial STC-1 expression in affected women. Endometrial biopsy samples were obtained from women with PCOS and controls, including samples from overweight/obese women with PCOS before and after a 3-month lifestyle intervention. A total of 98 PCOS and 85 control samples were used in immunohistochemistry, reverse-transcription polymerase chain reaction, or in vitro cell culture. STC-1 expression was analyzed at different cycle phases and in endometrial stromal cells (eSCs) after steroid hormone exposure. The eSCs were also challenged with 8-bromo-cAMP and hypoxia for STC-1 expression. The findings indicate that STC-1 expression is not steroid hormone mediated although secretory-phase STC-1 expression was blunted in PCOS. Lower expression seems to be related to attenuated STC-1 response to stressors in PCOS eSCs, shown as downregulation of protein kinase A activity. The 3-month lifestyle intervention did not restore STC-1 expression in PCOS endometrium. More studies are warranted to further elucidate the mechanisms behind the altered endometrial STC-1 expression and rescue mechanism in the PCOS endometrium.
Collapse
Affiliation(s)
- Masuma Khatun
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Riikka K Arffman
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Darja Lavogina
- Department of Bioorganic Chemistry, Institute of Chemistry, University of Tartu, Tartu, Estonia.,Competence Centre on Health Technologies, Tartu, Estonia
| | - Marika Kangasniemi
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Johanna Laru
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Anne Ahtikoski
- Department of Pathology, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Siri Lehtonen
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| | - Mariana Paulson
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Angelica Lindén Hirschberg
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Andres Salumets
- Competence Centre on Health Technologies, Tartu, Estonia.,Department of Obstetrics and Gynecology, Institute of Clinical Medicine, Tartu, Estonia.,Department of Biomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Leif C Andersson
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Terhi T Piltonen
- Department of Obstetrics and Gynecology, PEDEGO Research Unit, Medical Research Center, Oulu University Hospital, University of Oulu, Oulu, Finland
| |
Collapse
|
19
|
Song Y, Zhou F, Tan X, Liu X, Ding J, Zhang C, Li F, Zhu W, Ma W, Hu R, Zhang M. Bushen Huoxue recipe attenuates early pregnancy loss via activating endometrial COX2-PGE2 angiogenic signaling in mice. BMC Complement Med Ther 2021; 21:36. [PMID: 33446182 PMCID: PMC7809844 DOI: 10.1186/s12906-021-03201-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 01/04/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND During the fresh cycles of in vitro fertilization and embryo transfer, a disturbance in the reproductive endocrine environment following controlled ovarian hyperstimulation (COH) is closely related to compromised endometrial receptivity. This is a major disadvantage for women during pregnancy. Based on the theory of traditional Chinese medicine, Bushen Huoxue recipe (BSHXR) has been indicated to facilitate embryo implantation. METHODS The COH model (Kunming breed) was induced by injecting mice with pregnant mare serum gonadotrophin (0.4 IU/g) and human chorionic gonadotropin (1 IU/g), followed by treatment with BSHXR at three different concentrations (5.7, 11.4, and 22.8 g/kg), Bushen recipe (BSR) (5.7 g/kg), and Huoxue recipe (HXR) (5.7 g/kg). After successful mating, the pregnancy rate and implantation sites were examined on embryo day 8 (ED8), and the weight ratio of endometrium was calculated on ED4 midnight. Serum estrogen, progesterone, and endometrial PGE2 levels were measured using enzyme-linked immunosorbent assay. The endometrial microvasculature was evaluated using CD31 immunostaining. The protein and mRNA levels of the angiogenic factors in the endometrium were evaluated using western blot, immunohistochemistry, and polymerase chain reaction. RESULTS In the COH group, the pregnancy rate and implantation sites were significantly decreased, and abnormal serum hormone levels and impaired endometrial vascular development were observed. After BSHXR treatment, the supraphysiological serum progesterone level in COH mice was restored to normalcy. Moreover, the abnormal expression of the endometrial pro-angiogenic factors, including HIF1α, COX2-PGE2 pathway, and the down-stream factors, namely, MMP2, MMP9, TIMP2, and FGF2 after subjecting mice to COH was significantly improved after BSHXR treatment. CONCLUSION BSHXR could improve embryo implantation by regulating hormonal balance and modulating endometrial angiogenesis in mice, without inducing any side effects in normal pregnancy.
Collapse
Affiliation(s)
- Yufan Song
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Fanru Zhou
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Xiujuan Tan
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Xia Liu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Jiahui Ding
- Department of Obstetrics and Gynecology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Chu Zhang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Fan Li
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Wenxin Zhu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Wenwen Ma
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Runan Hu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Mingmin Zhang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
20
|
Cigarette Smoke Extract Activates Hypoxia-Inducible Factors in a Reactive Oxygen Species-Dependent Manner in Stroma Cells from Human Endometrium. Antioxidants (Basel) 2021; 10:antiox10010048. [PMID: 33401600 PMCID: PMC7823731 DOI: 10.3390/antiox10010048] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/31/2020] [Accepted: 12/31/2020] [Indexed: 02/07/2023] Open
Abstract
Cigarette smoking (CS) is a major contributing factor in the development of a large number of fatal and debilitating disorders, including degenerative diseases and cancers. Smoking and passive smoking also affect the establishment and maintenance of pregnancy. However, to the best of our knowledge, the effects of smoking on the human endometrium remain poorly understood. In this study, we investigated the regulatory mechanism underlying CS-induced hypoxia-inducible factor (HIF)-1α activation using primary human endometrial stromal cells and an immortalized cell line (KC02-44D). We found that the CS extract (CSE) increased reactive oxygen species levels and stimulated HIF-1α protein stabilization in endometrial stromal cells, and that CS-induced HIF-1α-dependent gene expression under non-hypoxic conditions in a concentration- and time-dependent manner. Additionally, we revealed the upregulated expression of a hypoxia-induced gene set following the CSE treatment, even under normoxic conditions. These results indicated that HIF-1α might play an important role in CS-exposure-induced cellular stress, inflammation, and endometrial remodeling.
Collapse
|
21
|
Kida N, Nishigaki A, Kakita‐Kobayashi M, Tsubokura H, Hashimoto Y, Yoshida A, Hisamatsu Y, Tsuzuki‐Nakao T, Murata H, Okada H. Exposure to cigarette smoke affects endometrial maturation including angiogenesis and decidualization. Reprod Med Biol 2021; 20:108-118. [PMID: 33488290 PMCID: PMC7812488 DOI: 10.1002/rmb2.12360] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/13/2020] [Accepted: 11/29/2020] [Indexed: 12/11/2022] Open
Abstract
PURPOSE To elucidate the effects of cigarette smoking on human endometrial maturation for reproductive function, the authors examined the in vitro effects of cigarette smoke extract (CSE) on angiogenesis and decidualization in primary human endometrial stromal cells (ESCs). METHODS Endometrial stromal cells were cultured with CSE and/or estradiol-17β (E2) and medroxyprogesterone acetate (MPA). The mRNA, protein levels, and protein secretion of the angiogenic factors and decidual specific factors were assessed using real-time polymerase chain reaction, Western blot analysis, and enzyme-linked immunosorbent assay, respectively. Decidualization was also monitored by the changes in cellular morphology. RESULTS Endometrial stromal cell proliferation substantially decreased after dose-dependent treatments with CSE at concentrations above 1%, whereas cell death was induced at treatment concentrations above 1% CSE. Treatments above 0.025% CSE led to increased vascular endothelial growth factor mRNA through hypoxia-inducible factor-1α accumulation. CSE concentrations at 0.01% and 0.025% increased the prolactin expression levels after treatment with E2 and MPA, whereas 0.1% and 0.25% CSE concentrations suppressed prolactin. Similar tendencies were observed in cellular morphology and other decidual specific factors. CONCLUSION These results suggest that exposure to cigarette smoke affects endometrial appropriate maturation including the processes of angiogenesis and decidualization in the reproductive system.
Collapse
Affiliation(s)
- Naoko Kida
- Department of Obstetrics and GynecologyKansai Medical UniversityOsakaJapan
| | - Akemi Nishigaki
- Department of Obstetrics and GynecologyKansai Medical UniversityOsakaJapan
| | | | - Hiroaki Tsubokura
- Department of Obstetrics and GynecologyKansai Medical UniversityOsakaJapan
| | - Yoshiko Hashimoto
- Department of Obstetrics and GynecologyKansai Medical UniversityOsakaJapan
| | - Aya Yoshida
- Department of Obstetrics and GynecologyKansai Medical UniversityOsakaJapan
| | - Yoji Hisamatsu
- Department of Obstetrics and GynecologyKansai Medical UniversityOsakaJapan
| | | | - Hiromi Murata
- Department of Obstetrics and GynecologyKansai Medical UniversityOsakaJapan
| | - Hidetaka Okada
- Department of Obstetrics and GynecologyKansai Medical UniversityOsakaJapan
| |
Collapse
|
22
|
Qu H, Khalil RA. Vascular mechanisms and molecular targets in hypertensive pregnancy and preeclampsia. Am J Physiol Heart Circ Physiol 2020; 319:H661-H681. [PMID: 32762557 DOI: 10.1152/ajpheart.00202.2020] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Preeclampsia is a major complication of pregnancy manifested as hypertension and often intrauterine growth restriction, but the underlying pathophysiological mechanisms are unclear. Predisposing genetic and environmental factors cause placental maladaptations leading to defective placentation, apoptosis of invasive cytotrophoblasts, inadequate expansive remodeling of the spiral arteries, reduced uteroplacental perfusion pressure, and placental ischemia. Placental ischemia promotes the release of bioactive factors into the maternal circulation, causing an imbalance between antiangiogenic soluble fms-like tyrosine kinase-1 and soluble endoglin and proangiogenic vascular endothelial growth factor, placental growth factor, and transforming growth factor-β. Placental ischemia also stimulates the release of proinflammatory cytokines, hypoxia-inducible factor, reactive oxygen species, and angiotensin type 1 receptor agonistic autoantibodies. These circulating factors target the vascular endothelium, causing generalized endotheliosis in systemic, renal, cerebral, and hepatic vessels, leading to decreases in endothelium-derived vasodilators such as nitric oxide, prostacyclin, and hyperpolarization factor and increases in vasoconstrictors such as endothelin-1 and thromboxane A2. The bioactive factors also target vascular smooth muscle and enhance the mechanisms of vascular contraction, including cytosolic Ca2+, protein kinase C, and Rho-kinase. The bioactive factors could also target matrix metalloproteinases and the extracellular matrix, causing inadequate vascular remodeling, increased arterial stiffening, and further increases in vascular resistance and hypertension. As therapeutic options are limited, understanding the underlying vascular mechanisms and molecular targets should help design new tools for the detection and management of hypertension in pregnancy and preeclampsia.
Collapse
Affiliation(s)
- Hongmei Qu
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
23
|
Narimani L, Boroujeni NB, Gholami M, Anbari K, Alavi SER, Ahmadi SAY, Boroujeni MB. Pre-Implantation Effects of Progesterone Administration on Ovarian Angiogenesis after Ovarian Stimulation: A Histological, Hormonal, and Molecular Analysis. JBRA Assist Reprod 2020; 24:289-295. [PMID: 32155017 PMCID: PMC7365533 DOI: 10.5935/1518-0557.20190076] [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: 05/01/2019] [Accepted: 11/10/2019] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVE Progesterone (P4) is known to directly affect ovarian tissue angiogenesis. The present study was designed to show how P4 affects ovarian angiogenesis in hormonal, histological, and molecular levels. METHODS Fifteen adult female NMRI mice were divided into three groups: Control Group; Case Group I (ovarian stimulation alone); and Case Group II (ovarian stimulation followed by P4 administration). Blood and ovarian tissue samples were assessed for hormonal, histological, and molecular alterations. Gene expression for ovarian vascular endothelium growth factor (VEGF) and hypoxia-inducible factor-1 alpha (HIF-1α) was analyzed using real-time PCR. RESULTS Ovarian hormone levels were increased in the case groups compared with the control group (p<0.05). Quantitative corpus luteum parameters were increased in the case groups compared with the control group (p<0.05). Quantitative ovarian vascular parameters were significantly different in the case groups compared with the control group. Gene expression analyses shows that the mice in Case Group I had higher levels of ovarian VEGF expression than the mice in the control group (p<0.05). No significant difference in gene expression was observed for HIF-1ɑ. CONCLUSION Treatment with P4 after ovarian stimulation enhanced ovarian angiogenesis by increasing hormone levels and causing significant structural changes.
Collapse
Affiliation(s)
- Leila Narimani
- Department of Anatomical Sciences, Lorestan University of Medical Sciences, Khorramabad, Iran
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Nasim Beigi Boroujeni
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mohammadreza Gholami
- Department of Anatomical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Khatereh Anbari
- Social Determinants of Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | | | - Seyyed Amir Yasin Ahmadi
- Pediatric Growth and Development Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Mandana Beigi Boroujeni
- Department of Anatomical Sciences, Lorestan University of Medical Sciences, Khorramabad, Iran
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
24
|
A hypoxia-induced Rab pathway regulates embryo implantation by controlled trafficking of secretory granules. Proc Natl Acad Sci U S A 2020; 117:14532-14542. [PMID: 32513733 DOI: 10.1073/pnas.2000810117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Implantation is initiated when an embryo attaches to the uterine luminal epithelium and subsequently penetrates into the underlying stroma to firmly embed in the endometrium. These events are followed by the formation of an extensive vascular network in the stroma that supports embryonic growth and ensures successful implantation. Interestingly, in many mammalian species, these processes of early pregnancy occur in a hypoxic environment. However, the mechanisms underlying maternal adaptation to hypoxia during early pregnancy remain unclear. In this study, using a knockout mouse model, we show that the transcription factor hypoxia-inducible factor 2 alpha (Hif2α), which is induced in subluminal stromal cells at the time of implantation, plays a crucial role during early pregnancy. Indeed, when preimplantation endometrial stromal cells are exposed to hypoxic conditions in vitro, we observed a striking enhancement in HIF2α expression. Further studies revealed that HIF2α regulates the expression of several metabolic and protein trafficking factors, including RAB27B, at the onset of implantation. RAB27B is a member of the Rab family of GTPases that allows controlled release of secretory granules. These granules are involved in trafficking MMP-9 from the stroma to the epithelium to promote luminal epithelial remodeling during embryo invasion. As pregnancy progresses, the HIF2α-RAB27B pathway additionally mediates crosstalk between stromal and endothelial cells via VEGF granules, developing the vascular network critical for establishing pregnancy. Collectively, our study provides insights into the intercellular communication mechanisms that operate during adaptation to hypoxia, which is essential for embryo implantation and establishment of pregnancy.
Collapse
|
25
|
Oliver KF, Geary TW, Kiser JN, Galliou JM, Van Emon ML, Seabury CM, Spencer TE, Neibergs HL. Loci associated with conception rate in crossbred beef heifers. PLoS One 2020; 15:e0230422. [PMID: 32271764 PMCID: PMC7145093 DOI: 10.1371/journal.pone.0230422] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 03/01/2020] [Indexed: 11/18/2022] Open
Abstract
The inability of beef cattle to maintain full term pregnancies has become an economic concern for the beef industry. Herd management and nutritional improvements have alleviated environmental impacts on embryonic and fetal loss, yet additional gains can be made through genomic selection. The objectives of this study were to identify loci and gene-sets in crossbred beef heifers associated with the number of services required to become pregnant (TBRD) and heifer conception rate at first service (HCR1). Heifers (n = 709) from a commercial beef operation underwent one round of artificial insemination, before exposure to bulls for natural service for 50 days. Pregnancy and time of conception was determined by ultrasound 35 days after the breeding season. Heifers were genotyped using the GeneSeek (Lincoln, NE) Bovine GGP50K BeadChip prior to genome-wide association analyses (GWAA) conducted using an EIGENSTRAT-like model to identify loci associated (P < 1 × 10−5) with TBRD and HCR1. One locus was associated (P = 8.97 × 10−6) with TBRD on BTA19 and included the positional candidate gene ASIC2, which is differentially expressed in the endometrium of fertility classified heifers, and the positional candidate gene, SPACA3. Gene-set enrichment analyses using SNP (GSEA-SNP) data, was performed and identified one gene-set, oxidoreductase activity, acting on paired donors, with incorporation or reduction of molecular oxygen as enriched (NES = 3.15) with TBRD and contained nine leading edge genes that contributed to the enrichment of the gene set. The enriched gene-set is involved in catalyzing oxidation-reduction reactions, which have been associated with oxidative stressors impacting pregnancy success. No loci were associated nor gene-sets enriched with HCR1. Identification of loci, positional candidate genes, gene-sets and leading edge genes enriched for fertility facilitate genomic selection that allows producers to select for reproductively superior cattle, reduce costs associated with infertility, and increase percent calf crop.
Collapse
Affiliation(s)
- K. F. Oliver
- Department of Animal Sciences, Washington State University, Pullman, WA, United States of America
| | - T. W. Geary
- USDA-ARS Fort Keogh LARRL, Miles City, MT, United States of America
| | - J. N. Kiser
- Department of Animal Sciences, Washington State University, Pullman, WA, United States of America
| | - J. M. Galliou
- Department of Animal Sciences, Washington State University, Pullman, WA, United States of America
| | - M. L. Van Emon
- Department of Animal and Range Sciences, Montana State University, Bozeman, MT, United States of America
| | - C. M. Seabury
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, TX, United States of America
| | - T. E. Spencer
- Division of Animal Sciences, University of Missouri, Columbia, MO, United States of America
| | - H. L. Neibergs
- Department of Animal and Range Sciences, Montana State University, Bozeman, MT, United States of America
- * E-mail:
| |
Collapse
|
26
|
Fukui Y, Hirota Y, Matsuo M, Gebril M, Akaeda S, Hiraoka T, Osuga Y. Uterine receptivity, embryo attachment, and embryo invasion: Multistep processes in embryo implantation. Reprod Med Biol 2019; 18:234-240. [PMID: 31312101 PMCID: PMC6613011 DOI: 10.1002/rmb2.12280] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 05/12/2019] [Accepted: 05/13/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Recurrent implantation failure is a critical issue in IVF-ET treatment. Successful embryo implantation needs appropriate molecular and cellular communications between embryo and uterus. Rodent models have been used intensively to understand these mechanisms. METHODS The molecular and cellular mechanisms of embryo implantation were described by referring to the previous literature investigated by us and others. The studies using mouse models of embryo implantation were mainly cited. RESULTS Progesterone (P4) produced by ovarian corpus luteum provides the uterus with receptivity to the embryo, and uterine epithelial growth arrest and stromal proliferation, what we call uterine proliferation-differentiation switching (PDS), take place in the peri-implantation period before embryo attachment. Uterine PDS is a hallmark of uterine receptivity, and several genes such as HAND2 and BMI1, control uterine PDS by modulating P4-PR signaling. As the next implantation process, embryo attachment onto the luminal epithelium occurs. This process is regulated by FOXA2-LIF pathway and planar cell polarity signaling. Then, the luminal epithelium at the embryo attachment site detaches from the stroma, which enables trophoblast invasion. This process of embryo invasion is regulated by HIF2α in the stroma. CONCLUSION These findings indicate that embryo implantation contains multistep processes regulated by specific molecular pathways.
Collapse
Affiliation(s)
- Yamato Fukui
- Department of Obstetrics and Gynecology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Yasushi Hirota
- Department of Obstetrics and Gynecology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Mitsunori Matsuo
- Department of Obstetrics and Gynecology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Mona Gebril
- Department of Obstetrics and Gynecology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Shun Akaeda
- Department of Obstetrics and Gynecology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Takehiro Hiraoka
- Department of Obstetrics and Gynecology, Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Graduate School of MedicineThe University of TokyoTokyoJapan
| |
Collapse
|
27
|
Abstract
Blastocyst implantation contains the following three processes: apposition, attachment, and invasion of the blastocyst. Ovarian hormone progesterone (P4) regulates these processes exquisitely. P4-induced molecular communications between the endometrial epithelium and stroma as well as endometrial proliferation-differentiation switching (PDS) until blastocyst attachment are fundamental steps in blastocyst implantation. Based on the knowledge obtained from the previous studies of mouse models by my group and others, this article outlines how P4 directs the uterus to complete blastocyst implantation.
Collapse
Affiliation(s)
- Yasushi Hirota
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
28
|
Yu W, Gao W, Rong D, Wu Z, Khalil RA. Molecular determinants of microvascular dysfunction in hypertensive pregnancy and preeclampsia. Microcirculation 2018; 26:e12508. [PMID: 30338879 PMCID: PMC6474836 DOI: 10.1111/micc.12508] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 10/05/2018] [Accepted: 10/15/2018] [Indexed: 12/16/2022]
Abstract
Preeclampsia is a pregnancy-related disorder characterized by hypertension and often fetal intrauterine growth restriction, but the underlying mechanisms are unclear. Defective placentation and apoptosis of invasive cytotrophoblasts cause inadequate remodeling of spiral arteries, placental ischemia, and reduced uterine perfusion pressure (RUPP). RUPP causes imbalance between the anti-angiogenic factors soluble fms-like tyrosine kinase-1 and soluble endoglin and the pro-angiogenic vascular endothelial growth factor and placental growth factor, and stimulates the release of proinflammatory cytokines, hypoxia-inducible factor, reactive oxygen species, and angiotensin AT1 receptor agonistic autoantibodies. These circulating factors target the vascular endothelium, smooth muscle and various components of the extracellular matrix. Generalized endotheliosis in systemic, renal, cerebral, and hepatic vessels causes decreases in endothelium-derived vasodilators such as nitric oxide, prostacyclin and hyperpolarization factor, and increases in vasoconstrictors such as endothelin-1 and thromboxane A2. Enhanced mechanisms of vascular smooth muscle contraction, such as intracellular Ca2+ , protein kinase C, and Rho-kinase cause further increases in vasoconstriction. Changes in matrix metalloproteinases and extracellular matrix cause inadequate vascular remodeling and increased arterial stiffening, leading to further increases in vascular resistance and hypertension. Therapeutic options are currently limited, but understanding the molecular determinants of microvascular dysfunction could help in the design of new approaches for the prediction and management of preeclampsia.
Collapse
Affiliation(s)
- Wentao Yu
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Wei Gao
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Dan Rong
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Zhixian Wu
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
29
|
Matsumoto L, Hirota Y, Saito-Fujita T, Takeda N, Tanaka T, Hiraoka T, Akaeda S, Fujita H, Shimizu-Hirota R, Igaue S, Matsuo M, Haraguchi H, Saito-Kanatani M, Fujii T, Osuga Y. HIF2α in the uterine stroma permits embryo invasion and luminal epithelium detachment. J Clin Invest 2018; 128:3186-3197. [PMID: 29911998 DOI: 10.1172/jci98931] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 05/01/2018] [Indexed: 01/21/2023] Open
Abstract
Although it has been reported that hypoxia inducible factor 2 α (Hif2a), a major transcriptional factor inducible by low oxygen tension, is expressed in the mouse uterus during embryo implantation, its role in pregnancy outcomes remains unclear. This study aimed to clarify functions of uterine HIF using transgenic mouse models. Mice with deletion of Hif2a in the whole uterus (Hif2a-uKO mice) showed infertility due to implantation failure. Supplementation with progesterone (P4) and leukemia inhibitory factor (LIF) restored decidual growth arrest and aberrant position of implantation sites in Hif2a-uKO mice, respectively, but did not rescue pregnancy failure. Histological analyses in Hif2a-uKO mice revealed persistence of the intact luminal epithelium, which blocked direct contact between stroma and embryo, inactivation of PI3K-AKT pathway (embryonic survival signal), and failed embryo invasion. Mice with stromal deletion of Hif2a (Hif2a-sKO mice) showed infertility with impaired embryo invasion and those with epithelial deletion of Hif2a (Hif2a-eKO mice) showed normal fertility, suggesting the importance of stromal HIF2α in embryo invasion. This was reflected in reduced expression of membrane type 2 metalloproteinase (MT2-MMP), lysyl oxidase (LOX), VEGF, and adrenomedullin (ADM) in Hif2a-uKO stroma at the attachment site, suggesting that stromal HIF2α regulates these mediators to support blastocyst invasion. These findings provide new insight that stromal HIF2α allows trophoblast invasion through detachment of the luminal epithelium and activation of an embryonic survival signal.
Collapse
Affiliation(s)
- Leona Matsumoto
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yasushi Hirota
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.,Precursory Research for Innovative Medical Care (PRIME), Japan Agency for Medical Research and Development (AMED), Bunkyo-ku, Tokyo, Japan
| | - Tomoko Saito-Fujita
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Norihiko Takeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Tomoki Tanaka
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Takehiro Hiraoka
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Shun Akaeda
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hidetoshi Fujita
- Department of Future Medical Science, Institute of Medical Science, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Ryoko Shimizu-Hirota
- Department of Internal Medicine, Center of Preventive Medicine, School of Medicine, Keio University, Shinjuku-ku, Tokyo, Japan
| | - Shota Igaue
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Mitsunori Matsuo
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hirofumi Haraguchi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Mayuko Saito-Kanatani
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Tomoyuki Fujii
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
30
|
Soares MJ, Iqbal K, Kozai K. Hypoxia and Placental Development. Birth Defects Res 2018; 109:1309-1329. [PMID: 29105383 DOI: 10.1002/bdr2.1135] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 09/04/2017] [Indexed: 12/17/2022]
Abstract
Hemochorial placentation is orchestrated through highly regulated temporal and spatial decisions governing the fate of trophoblast stem/progenitor cells. Trophoblast cell acquisition of specializations facilitating invasion and uterine spiral artery remodeling is a labile process, sensitive to the environment, and represents a process that is vulnerable to dysmorphogenesis in pathologic states. Hypoxia is a signal guiding placental development, and molecular mechanisms directing cellular adaptations to low oxygen tension are integral to trophoblast cell differentiation and placentation. Hypoxia can also be used as an experimental tool to investigate regulatory processes controlling hemochorial placentation. These developmental processes are conserved in mouse, rat, and human placentation. Consequently, elements of these developmental events can be modeled and hypotheses tested in trophoblast stem cells and in genetically manipulated rodents. Hypoxia is also a consequence of a failed placenta, yielding pathologies that can adversely affect maternal adjustments to pregnancy, fetal health, and susceptibility to adult disease. The capacity of the placenta for adaptation to environmental challenges highlights the importance of its plasticity in safeguarding a healthy pregnancy. Birth Defects Research 109:1309-1329, 2017.© 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Michael J Soares
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas.,Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas.,Fetal Health Research, Children's Research Institute, Children's Mercy, Kansas City, Missouri
| | - Khursheed Iqbal
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Keisuke Kozai
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
31
|
Sones JL, Merriam AA, Seffens A, Brown-Grant DA, Butler SD, Zhao AM, Xu X, Shawber CJ, Grenier JK, Douglas NC. Angiogenic factor imbalance precedes complement deposition in placentae of the BPH/5 model of preeclampsia. FASEB J 2018; 32:2574-2586. [PMID: 29279353 DOI: 10.1096/fj.201701008r] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Preeclampsia (PE), a hypertensive disorder of pregnancy, is a leading cause of maternal and fetal morbidity and mortality. Although the etiology is unknown, PE is thought to be caused by defective implantation and decidualization in pregnancy. Pregnant blood pressure high (BPH)/5 mice spontaneously develop placentopathies and maternal features of human PE. We hypothesized that BPH/5 implantation sites have transcriptomic alterations. Next-generation RNA sequencing of implantation sites at peak decidualization, embryonic day (E)7.5, revealed complement gene up-regulation in BPH/5 vs. controls. In BPH/5, expression of complement factor 3 was increased around the decidual vasculature of E7.5 implantation sites and in the trophoblast giant cell layer of E10.5 placentae. Altered expression of VEGF pathway genes in E5.5 BPH/5 implantation sites preceded complement dysregulation, which correlated with abnormal vasculature and increased placental growth factor mRNA and VEGF164 expression at E7.5. By E10.5, proangiogenic genes were down-regulated, whereas antiangiogenic sFlt-1 was up-regulated in BPH/5 placentae. We found that early local misexpression of VEGF genes and abnormal decidual vasculature preceded sFlt-1 overexpression and increased complement deposition in BPH/5 placentae. Our findings suggest that abnormal decidual angiogenesis precedes complement activation, which in turn contributes to the aberrant trophoblast invasion and poor placentation that underlie PE.-Sones, J. L., Merriam, A. A., Seffens, A., Brown-Grant, D.-A., Butler, S. D., Zhao, A. M., Xu, X., Shawber, C. J., Grenier, J. K., Douglas, N. C. Angiogenic factor imbalance precedes complement deposition in placentae of the BPH/5 model of preeclampsia.
Collapse
Affiliation(s)
- Jennifer L Sones
- Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Audrey A Merriam
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Angelina Seffens
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Dex-Ann Brown-Grant
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Scott D Butler
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA; and
| | - Anna M Zhao
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Xinjing Xu
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Carrie J Shawber
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Jennifer K Grenier
- RNA Sequencing Core, Center for Reproductive Genomics, Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Nataki C Douglas
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, Columbia University College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
32
|
Kwiatkowska J, Wąsowska B, Gilun P. Expression of hypoxia inducible factor 1α and antioxidant enzymes: Superoxide dismutases-1 and -2 in ischemic porcine endometrium. Reprod Biol 2017. [PMID: 28624353 DOI: 10.1016/j.repbio.2017.05.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The aim of this study was to determine how 60-min ischemia changes the expression of hypoxia inducible factor 1α (HIF-1α) and superoxide dismutases (SOD)-1 and -2 in selected regions of porcine uterine horns. The results showed that 60-min ischemia of the porcine uterus conducted at the mid-secretory estrous phase caused decreased HIF-1α and increased SOD-2 gene expression. Higher expression of SOD-2 suggests that this enzyme may play an important role in the suppression of HIF-1α accumulation in an ischemic endometrium.
Collapse
Affiliation(s)
- Joanna Kwiatkowska
- Department of Local Physiological Regulations, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748 Olsztyn, Poland
| | - Barbara Wąsowska
- Department of Local Physiological Regulations, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748 Olsztyn, Poland.
| | - Przemysław Gilun
- Department of Local Physiological Regulations, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748 Olsztyn, Poland
| |
Collapse
|
33
|
Chen J, Khalil RA. Matrix Metalloproteinases in Normal Pregnancy and Preeclampsia. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 148:87-165. [PMID: 28662830 PMCID: PMC5548443 DOI: 10.1016/bs.pmbts.2017.04.001] [Citation(s) in RCA: 183] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Normal pregnancy is associated with marked hemodynamic and uterine changes that allow adequate uteroplacental blood flow and uterine expansion for the growing fetus. These pregnancy-associated changes involve significant uteroplacental and vascular remodeling. Matrix metalloproteinases (MMPs) are important regulators of vascular and uterine remodeling. Increases in MMP-2 and MMP-9 have been implicated in vasodilation, placentation, and uterine expansion during normal pregnancy. The increases in MMPs could be induced by the increased production of estrogen and progesterone during pregnancy. MMP expression/activity may be altered during complications of pregnancy. Decreased vascular MMP-2 and MMP-9 may lead to decreased vasodilation, increased vasoconstriction, hypertensive pregnancy, and preeclampsia. Abnormal expression of uteroplacental integrins, cytokines, and MMPs may lead to decreased maternal tolerance, apoptosis of invasive trophoblast cells, inadequate remodeling of spiral arteries, and reduced uterine perfusion pressure (RUPP). RUPP may cause imbalance between the antiangiogenic factors soluble fms-like tyrosine kinase-1 and soluble endoglin and the proangiogenic vascular endothelial growth factor and placental growth factor, or stimulate the release of inflammatory cytokines, hypoxia-inducible factor, reactive oxygen species, and angiotensin AT1 receptor agonistic autoantibodies. These circulating factors could target MMPs in the extracellular matrix as well as endothelial and vascular smooth muscle cells, causing generalized vascular dysfunction, increased vasoconstriction and hypertension in pregnancy. MMP activity can also be altered by endogenous tissue inhibitors of metalloproteinases (TIMPs) and changes in the MMP/TIMP ratio. In addition to their vascular effects, decreases in expression/activity of MMP-2 and MMP-9 in the uterus could impede uterine growth and expansion and lead to premature labor. Understanding the role of MMPs in uteroplacental and vascular remodeling and function could help design new approaches for prediction and management of preeclampsia and premature labor.
Collapse
Affiliation(s)
- Juanjuan Chen
- Vascular Surgery Research Laboratories, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States
| | - Raouf A Khalil
- Vascular Surgery Research Laboratories, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.
| |
Collapse
|
34
|
Liu JL, Zhao M, Peng Y, Fu YS. Identification of gene expression changes in rabbit uterus during embryo implantation. Genomics 2016; 107:216-21. [PMID: 27071951 DOI: 10.1016/j.ygeno.2016.03.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 03/06/2016] [Accepted: 03/29/2016] [Indexed: 11/30/2022]
Abstract
Embryo implantation in the rabbit is unique in that a typical fusion type of implantation is employed, in which trophoblast cells adhere and fuse to the apical surface of uterine epithelial cells. In the present study, we analyzed global gene expression changes in the rabbit uterus during embryo implantation by using RNA-seq. We identified a total of 536 differentially expressed genes (fold change >2 and adjusted p-value <0.01), of which 266 genes were down-regulated and 270 genes were up-regulated at the implantation site compared to the inter-implantation site. Functional clustering revealed that cell adhesion is among top ranked enriched terms from both gene ontology and pathway analysis, highlighting the importance of cell adhesion during embryo implantation in rabbits. Through gene network analysis, we prioritized 9 genes using the hub gene method. Our study provides a valuable resource for in-depth understanding of the mechanism underlying embryo implantation in rabbits.
Collapse
Affiliation(s)
- Ji-Long Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.
| | - Miao Zhao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Ying Peng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yong-Sheng Fu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| |
Collapse
|
35
|
Marshall SA, Ng L, Unemori EN, Girling JE, Parry LJ. Relaxin deficiency results in increased expression of angiogenesis- and remodelling-related genes in the uterus of early pregnant mice but does not affect endometrial angiogenesis prior to implantation. Reprod Biol Endocrinol 2016; 14:11. [PMID: 27005936 PMCID: PMC4802869 DOI: 10.1186/s12958-016-0148-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 03/14/2016] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Extensive uterine adaptations, including angiogenesis, occur prior to implantation in early pregnancy and are potentially regulated by the peptide hormone relaxin. This was investigated in two studies. First, we took a microarray approach using human endometrial stromal (HES) cells treated with relaxin in vitro to screen for target genes. Then we aimed to investigate whether or not relaxin deficiency in mice affected uterine expression of representative genes associated with angiogenesis and uterine remodeling, and also blood vessel proliferation in the pre-implantation mouse endometrium. METHODS Normal HES cells were isolated and treated with recombinant human relaxin (10 ng/ml) for 24 h before microarray analysis. Reverse transcriptase PCR was used to analyze gene expression of relaxin and its receptor (Rxfp1) in ovaries and uteri; quantitative PCR was used to analyze steroid receptor, angiogenesis and extracellular matrix remodeling genes in the uteri of wild type (Rln+/+) and Rln-/- mice on days 1-4 of pregnancy. Immunohistochemistry localized endometrial endothelial cell proliferation and mass spectrometry measured steroid hormones in the plasma. RESULTS Microarray analysis identified 63 well-characterized genes that were differentially regulated in HES cells after relaxin treatment. Expression of some of these genes was increased in the uterus of Rln+/+ mice by day 4 of pregnancy. There was significantly higher vascular endothelial growth factor A (VegfA), estrogen receptor 1 (Esr1), progesterone receptor (Pgr), Rxfp1, egl-9 family hypoxia-inducible factor 1 (Egln1), hypoxia inducible factor 1 alpha (Hif1α), matrix metalloproteinase 14 (Mmp14) and ankryn repeat domain 37 (Ankrd37) in Rln-/- compared to Rln+/+ mice on day 1. Progesterone receptor expression and plasma progesterone levels were higher in Rln-/- mice compared to Rln+/+ mice. However, endometrial angiogenesis was not advanced as pre-implantation endothelial cell proliferation did not differ between genotypes. CONCLUSIONS Relaxin treatment modulates expression of a variety of angiogenesis-related genes in HES cells. However, despite accelerated uterine gene expression of steroid receptor, progesterone and angiogenesis and extracellular matrix remodeling genes in Rln-/- mice, there was no impact on angiogenesis. We conclude that although relaxin deficiency results in phenotypic changes in the pre-implantation uterus, endogenous relaxin does not play a major role in pre-implantation angiogenesis in the mouse uterus.
Collapse
Affiliation(s)
- Sarah A. Marshall
- School of BioSciences, The University of Melbourne, Royal Parade, Parkville, Victoria Australia
| | - Leelee Ng
- School of BioSciences, The University of Melbourne, Royal Parade, Parkville, Victoria Australia
| | | | - Jane E. Girling
- Gynaecology Research Centre, Department of Obstetrics and Gynecology, The University of Melbourne and Royal Women’s Hospital, Parkville, Victoria Australia
| | - Laura J. Parry
- School of BioSciences, The University of Melbourne, Royal Parade, Parkville, Victoria Australia
| |
Collapse
|
36
|
Expression of hypoxia-inducible factor-1 by trophectoderm cells in response to hypoxia and epidermal growth factor. Biochem Biophys Res Commun 2016; 469:176-82. [DOI: 10.1016/j.bbrc.2015.11.091] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 11/20/2015] [Indexed: 11/18/2022]
|
37
|
Zuo RJ, Gu XW, Qi QR, Wang TS, Zhao XY, Liu JL, Yang ZM. Warburg-like Glycolysis and Lactate Shuttle in Mouse Decidua during Early Pregnancy. J Biol Chem 2015; 290:21280-91. [PMID: 26178372 DOI: 10.1074/jbc.m115.656629] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Indexed: 12/15/2022] Open
Abstract
Decidualization is an essential process of maternal endometrial stromal cells to support pregnancy. Although it is known that enhanced glucose influx is critical for decidualization, the underlying mechanism in regulating glucose metabolism in decidua remains insufficiently understood. Here, we demonstrate that aerobic glycolysis-related genes and factors are all substantially induced during decidualization, indicating the existence of Warburg-like glycolysis in decidua. In vitro, progesterone activates hypoxia-inducible factor 1α (Hif1α) and c-Myc through Pi3k-Akt signaling pathway to maintain aerobic glycolysis in decidualizing cells. Knocking down of pyruvate kinase M2 (Pkm2) attenuates the induction of decidual marker gene. Decidual formation in vivo is also impaired by glycolysis inhibitor 3-bromopyruvate. Besides, lactate exporter monocarboxylate transporter 4 (Mct4) is induced in newly formed decidual cells, whereas lactate importer Mct1 and proliferation marker Ki-67 are complementarily located in the surrounding undifferentiated cells, which are supposed to consume lactate for proliferation. Hif1α activation is required for lactate-dependent proliferation of the undifferentiated cells. Inhibition of lactate flux leads to compromised decidualization and decelerated lactate-dependent proliferation. In summary, we reveal that Warburg-like glycolysis and local lactate shuttle are activated in decidua and play important roles for supporting early pregnancy.
Collapse
Affiliation(s)
- Ru-Juan Zuo
- From the College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642 and
| | - Xiao-Wei Gu
- the Department of Biology, Shantou University, Shantou 515063, China
| | - Qian-Rong Qi
- the Department of Biology, Shantou University, Shantou 515063, China
| | - Tong-Song Wang
- the Department of Biology, Shantou University, Shantou 515063, China
| | - Xu-Yu Zhao
- the Department of Biology, Shantou University, Shantou 515063, China
| | - Ji-Long Liu
- From the College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642 and
| | - Zeng-Ming Yang
- From the College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642 and
| |
Collapse
|
38
|
Chen X, Liu J, He B, Li Y, Liu S, Wu B, Wang S, Zhang S, Xu X, Wang J. Vascular endothelial growth factor (VEGF) regulation by hypoxia inducible factor-1 alpha (HIF1A) starts and peaks during endometrial breakdown, not repair, in a mouse menstrual-like model. Hum Reprod 2015; 30:2160-70. [DOI: 10.1093/humrep/dev156] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 06/05/2015] [Indexed: 12/23/2022] Open
|
39
|
Tang X, Chen Y, Ran H, Jiang Y, He B, Wang B, Kong S, Wang H. Systemic morphine treatment derails normal uterine receptivity, leading to embryo implantation failure in mice. Biol Reprod 2015; 92:118. [PMID: 25855262 DOI: 10.1095/biolreprod.115.128686] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 04/02/2015] [Indexed: 11/01/2022] Open
Abstract
Morphine is the oldest worldwide well-known opioid agonist used for pain treatment in clinic, and its illicit use is often associated with adverse pregnancy outcomes in humans. Because of recent dramatic increases in nonmedicinal morphine abuse, one emerging issue is the further revelation of the dark side of illicit opioid uses, particularly in early pregnancy events. In this respect, we have demonstrated that opioid signaling is functionally operative during preimplantation embryo development in mice. However, the pathophysiological significance of the opioid system on uterine functions at peri-implantation remained elusive. In the present study, we demonstrated that opioid receptors were spatiotemporally expressed in the uterus during the peri-implantation period. Employing a pharmacological approach combined with embryo transfer experiments, we further observed that although systemic morphine treatment exerts no apparent adverse influence on preimplantation ovarian secretion of progesterone and estrogen, this aberrant activation of opioid signaling by morphine induces impaired luminal epithelial differentiation, decreased stromal cell proliferation, and poor angiogenesis, and thus hampers uterine receptivity and embryo implantation. These novel findings add a new line of evidence to better understand the causes for obvious adverse effects of opioid abuse on pregnancy success in women.
Collapse
Affiliation(s)
- Xiaofang Tang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, PR China University of Chinese Academy of Sciences, Beijing, PR China
| | - Yongjie Chen
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, PR China University of Chinese Academy of Sciences, Beijing, PR China
| | - Hao Ran
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, PR China
| | - Yufei Jiang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, PR China
| | - Bo He
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, PR China University of Chinese Academy of Sciences, Beijing, PR China
| | - Bingyan Wang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, PR China
| | - Shuangbo Kong
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, PR China
| | - Haibin Wang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, PR China
| |
Collapse
|
40
|
Expression of hypoxia-inducible factors and vascular endothelial growth factor during pregnancy in the feline uterus. Theriogenology 2015; 84:24-33. [PMID: 25794839 DOI: 10.1016/j.theriogenology.2015.02.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 02/03/2015] [Accepted: 02/05/2015] [Indexed: 12/23/2022]
Abstract
Hypoxia-inducible factors (HIFs) and vascular endothelial growth factor (VEGF) have critical roles during the development of the fetomaternal unit. The HIFs regulate placentation and vascularization by stimulation of VEGF gene expression. This study aimed to investigate the expression profiles of HIF gene family and VEGF in the cat uterus during pregnancy. Tissue samples of the whole uterine wall were collected after ovariohysterectomy and allocated to the following groups: embryo positive (group 1 [G1], n = 7, 7 days after mating), early pregnancy (group 2 [G2], n = 7, 20 days after mating), mid-pregnancy (group 3 [G3], n = 7, 24 days after mating), late pregnancy (group 4 [G4], n = 7, 30-45 days after mating), and oocyte positive groups (group 5 [G5], n = 7, 7 days after induction of ovulation with GnRH analog). Relative mRNA levels were determined by real-time polymerase chain reaction. As housekeeping gene, glyceraldehyde-3-phosphate dehydrogenase was used. The relative gene expression of HIF1A in G5 was found to be significantly higher than that of other groups (G1, G2, G3, and G4) (P < 0.05). In addition, the expression of HIF2A in G5 was higher than that of G1 and HIF2A gene expression at placentation sites of G4 was higher than in G1, G2, and G3 (P < 0.05). Immunohistochemistry indicated that HIF1A, HIF2A, and VEGF expressions were observed in different cell types of uterine and placental tissues in late pregnancy and oocyte groups. The expression of HIF3A did not change significantly in any group investigated. These observations suggest that HIFs and VEGF may play a role in the establishment and development of pregnancy.
Collapse
|
41
|
First trimester human placental factors induce breast cancer cell autophagy. Breast Cancer Res Treat 2015; 149:645-54. [DOI: 10.1007/s10549-015-3266-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 12/23/2014] [Indexed: 12/20/2022]
|
42
|
Xie Y, Li Y, Kong Y. OPN induces FoxM1 expression and localization through ERK 1/2, AKT, and p38 signaling pathway in HEC-1A cells. Int J Mol Sci 2014; 15:23345-58. [PMID: 25522167 PMCID: PMC4284770 DOI: 10.3390/ijms151223345] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 11/25/2014] [Accepted: 12/11/2014] [Indexed: 01/05/2023] Open
Abstract
Mammalian embryo implantation is an extremely complex process and requires endometrial receptivity. In order to establish this receptivity, sequential proliferation and differentiation during the menstrual cycle is necessary. Forkhead box M1 (FoxM1) is described as a major oncogenic transcription factor in tumor initiation, promotion and progression. According to these functions, we believe that FoxM1 should also play an essential role in embryo implantation. Osteopontin (OPN), an adhesion molecule, has been studied extensively in reproduction. In this study, we observed the expression and distribution of FoxM1 during the proliferative-phase and secretory-phase human endometrium and the pre-implantation mouse uterus firstly. Then we observed the relationship between OPN and FoxM1. Our results showed that FoxM1 was mainly distributed in glandular epithelium. OPN increased the expression of FoxM1 in the human uterine epithelial cell line HEC-1A cells in a time- and concentration-dependent manner. OPN regulates FoxM1 to influence HEC-1A cell proliferation through extracellular regulated protein kinases (ERK 1/2), protein kinase B (PKB, AKT), and the p38 mitogen activated protein kinases (p38MAPK, p38) signaling pathway. Inhibition of ERK 1/2, AKT and p38 suppressed OPN-induced FoxM1 expression and location. Our data indicate that FoxM1 might be regulated by OPN to influence endometrial proliferation to establish endometrial receptivity.
Collapse
Affiliation(s)
- Yunpeng Xie
- Department of Biochemistry and Molecular, Dalian Medical University, Dalian 116044, China.
| | - Yinghua Li
- Department of Biochemistry and Molecular, Dalian Medical University, Dalian 116044, China.
| | - Ying Kong
- Department of Biochemistry and Molecular, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
43
|
Shin H, Choi S, Lim HJ. Relationship between reactive oxygen species and autophagy in dormant mouse blastocysts during delayed implantation. Clin Exp Reprod Med 2014; 41:125-31. [PMID: 25309857 PMCID: PMC4192453 DOI: 10.5653/cerm.2014.41.3.125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 08/19/2014] [Accepted: 09/15/2014] [Indexed: 12/12/2022] Open
Abstract
Objective Under estrogen deficiency, blastocysts cannot initiate implantation and enter dormancy. Dormant blastocysts live longer in utero than normal blastocysts, and autophagy has been suggested as a mechanism underlying the sustained survival of dormant blastocysts during delayed implantation. Autophagy is a cellular degradation pathway and a central component of the integrated stress response. Reactive oxygen species (ROS) are produced within cells during normal metabolism, but their levels increase dramatically under stressful conditions. We investigated whether heightened autophagy in dormant blastocysts is associated with the increased oxidative stress under the unfavorable condition of delayed implantation. Methods To visualize ROS production, day 8 (short-term dormancy) and day 20 (long-term dormancy) dormant blastocysts were loaded with 1-µM 5-(and-6)-chloromethyl-2', 7'-dichlorodihydrofluorescein diacetate, acetyl ester (CM-H2DCFDA). To block autophagic activation, 3-methyladenine (3-MA) and wortmannin were used in vivo and in vitro, respectively. Results We observed that ROS production was not significantly affected by the status of dormancy; in other words, both dormant and activated blastocysts showed high levels of ROS. However, ROS production was higher in the dormant blastocysts of the long-term dormancy group than in those of the short-term group. The addition of wortmannin to dormant blastocysts in vitro and 3-MA injection in vivo significantly increased ROS production in the short-term dormant blastocysts. In the long-term dormant blastocysts, ROS levels were not significantly affected by the treatment of the autophagy inhibitor. Conclusion During delayed implantation, heightened autophagy in dormant blastocysts may be operative as a potential mechanism to reduce oxidative stress. Further, ROS may be one of the potential causes of compromised developmental competence of long-term dormant blastocysts after implantation.
Collapse
Affiliation(s)
- Hyejin Shin
- Department of Biomedical Science and Technology, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea
| | - Soyoung Choi
- Department of Biomedical Science and Technology, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea
| | - Hyunjung Jade Lim
- Department of Biomedical Science and Technology, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea. ; Department of Veterinary Medicine, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea
| |
Collapse
|
44
|
Lima PDA, Zhang J, Dunk C, Lye SJ, Croy BA. Leukocyte driven-decidual angiogenesis in early pregnancy. Cell Mol Immunol 2014; 11:522-37. [PMID: 25066422 DOI: 10.1038/cmi.2014.63] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 06/21/2014] [Accepted: 06/22/2014] [Indexed: 12/15/2022] Open
Abstract
Successful pregnancy and long-term, post-natal maternal and offspring cardiac, vascular and metabolic health require key maternal cardiovascular adaptations over gestation. Within the pregnant decidualizing uterus, coordinated vascular, immunological and stromal cell changes occur. Considerable attention has been given to the roles of uterine natural killer (uNK) cells in initiating decidual spiral arterial remodeling, a process normally completed by mid-gestation in mice and in humans. However, leukocyte roles in much earlier, region specific, decidual vascular remodeling are now being defined. Interest in immune cell-promoted vascular remodeling is driven by vascular aberrations that are reported in human gestational complications such as infertility, recurrent spontaneous abortion, preeclampsia (PE) and fetal growth restriction. Appropriate maternal cardiovascular responses during pregnancy protect mothers and their children from later cardiovascular disease risk elevation. One of the earliest uterine responses to pregnancy in species with hemochorial placentation is stromal cell decidualization, which creates unique niches for angiogenesis and leukocyte recruitment. In early decidua basalis, the aspect of the implantation site that will cradle the developing placenta and provide the major blood vessels to support mature placental functions, leukocytes are greatly enriched and display specialized properties. UNK cells, the most abundant leukocyte subset in early decidua basalis, have angiogenic abilities and are essential for normal early decidual angiogenesis. The regulation of uNK cells and their roles in determining maternal and progeny cardiovascular health over pregnancy and postpartum are discussed.
Collapse
Affiliation(s)
- Patricia D A Lima
- Ottawa Hospital Research Institute, The Ottawa Hospital General Campus, Critical Care Wing, Ottawa, ON, Canada
| | - Jianhong Zhang
- Research Centre for Women's and Infants' Health, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Caroline Dunk
- Research Centre for Women's and Infants' Health, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Stephen J Lye
- 1] Research Centre for Women's and Infants' Health, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada [2] Department of Physiology and University of Toronto, Toronto, ON, Canada [3] Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON, Canada
| | - B Anne Croy
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| |
Collapse
|
45
|
Yoshii A, Kitahara S, Ueta H, Matsuno K, Ezaki T. Role of uterine contraction in regeneration of the murine postpartum endometrium. Biol Reprod 2014; 91:32. [PMID: 24966392 DOI: 10.1095/biolreprod.114.117929] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The endometrium undergoes continuous repair and regeneration without scarring throughout the reproductive life of women. However, the mechanisms responsible for this complete restoration remain mostly unexplored. We hypothesized that the ischemic state and local hypoxia present after parturition may create a special microenvironment for endometrial healing, and that this ischemia might be caused by reduction in organ volume via postpartum uterine contraction. Here, we developed a mouse model using a combination of cesarean section and the administration of a beta 2 adrenergic receptor agonist (ritodrine hydrochloride) in postpartum mice that had been ovariectomized to exclude the effect of ovarian hormones. Our results revealed that transient hypoxia indeed occurred in postpartum uteri. Furthermore, we found that the number of M2 macrophages, which play a central role in wound healing, peaked on Postpartum Day 3 and gradually decreased thereafter in hypoxic injury sites. Almost concurrently, significant upregulation of vascular endothelial growth factor and transforming growth factor beta (TGFbeta) was observed. In particular, the antifibrotic factor TGFbeta3 was released during the endometrial healing process. These changes were significantly suppressed by inhibition of uterine contraction. Taken together, these results suggest that uterine contraction is essential, not only for hemostasis, but also for endometrial regeneration, leading to a process that involves the activation of macrophages, increased endometrial cell proliferation, and upregulation of nonfibrotic growth factors. This study paves the way to a novel approach for investigating the process of scarless wound healing.
Collapse
Affiliation(s)
- Asuka Yoshii
- Department of Anatomy and Developmental Biology, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Shuji Kitahara
- Department of Anatomy and Developmental Biology, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Hisashi Ueta
- Department of Anatomy (Macro), Dokkyo Medical University, Tochigi, Japan
| | - Kenjiro Matsuno
- Department of Anatomy (Macro), Dokkyo Medical University, Tochigi, Japan
| | - Taichi Ezaki
- Department of Anatomy and Developmental Biology, School of Medicine, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
46
|
Zuo RJ, Zhao YC, Lei W, Wang TS, Wang BC, Yang ZM. Crystallin αB acts as a molecular guard in mouse decidualization: regulation and function during early pregnancy. FEBS Lett 2014; 588:2944-51. [PMID: 24951838 DOI: 10.1016/j.febslet.2014.05.045] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Revised: 05/08/2014] [Accepted: 05/28/2014] [Indexed: 02/06/2023]
Abstract
Although decidualization is crucial for the establishment of successful pregnancy, the molecular mechanism underlying decidualization remains poorly understood. Crystallin αB (CryAB), a small heat shock protein (sHSP), is up-regulated and phosphorylated in mouse decidua. In mouse primary endometrial stromal cells, CryAB is induced upon progesterone treatment via HIF1α. In addition, CryAB is strongly phosphorylated through the p38-MAPK pathway under stress or during in vitro decidualization. Knockdown of CryAB results in the increase of apoptosis of stromal cells and inhibits decidualization under oxidative or inflammatory stress. Our data indicate that CryAB protects decidualization against stress conditions.
Collapse
Affiliation(s)
- Ru-Juan Zuo
- School of Life Science, Xiamen University, Xiamen 361005, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Yue-Chao Zhao
- School of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Wei Lei
- School of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Tong-Song Wang
- School of Science, Shantou University, Shantou 515063, China
| | - Bao-Cheng Wang
- School of Science, Shantou University, Shantou 515063, China
| | - Zeng-Ming Yang
- School of Life Science, Xiamen University, Xiamen 361005, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
47
|
Xu X, Leng JY, Gao F, Zhao ZA, Deng WB, Liang XH, Zhang YJ, Zhang ZR, Li M, Sha AG, Yang ZM. Differential expression and anti-oxidant function of glutathione peroxidase 3 in mouse uterus during decidualization. FEBS Lett 2014; 588:1580-9. [PMID: 24631040 DOI: 10.1016/j.febslet.2014.02.043] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 02/17/2014] [Accepted: 02/20/2014] [Indexed: 01/13/2023]
Abstract
Glutathione peroxidase 3 (GPX3) is an important member of antioxidant enzymes for reducing reactive oxygen species and maintaining the oxygen balance. Gpx3 mRNA is strongly expressed in decidual cells from days 5 to 8 of pregnancy. After pregnant mice are treated with GPX inhibitor for 3 days, pregnancy rate is significantly reduced. Progesterone stimulates Gpx3 expression through PR/HIF1α in mouse endometrial stromal cells. In the decidua, the high level of GPX3 expression is closely associated with the reduction of hydrogen peroxide (H2O2). Based on our data, GPX3 may play a major role in reducing H2O2 during decidualization.
Collapse
Affiliation(s)
- Xiu Xu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; School of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Jing-Yu Leng
- School of Life Science, Xiamen University, Xiamen 361005, China
| | - Fei Gao
- School of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Zhen-Ao Zhao
- School of Life Science, Northeast Agricultural University, Harbin 150030, China
| | - Wen-Bo Deng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Xiao-Huan Liang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Yi-Juan Zhang
- School of Life Science, Xiamen University, Xiamen 361005, China
| | - Zhi-Rong Zhang
- School of Life Science, Xiamen University, Xiamen 361005, China
| | - Ming Li
- School of Life Science, Xiamen University, Xiamen 361005, China
| | - Ai-Guo Sha
- Reproductive Medicine Center, Bailu Hospital, Xiamen 361000, China
| | - Zeng-Ming Yang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
48
|
Ji Y, Lu X, Zhong Q, Liu P, An Y, Zhang Y, Zhang S, Jia R, Tesfamariam IG, Kahsay AG, Zhang L, Zhu W, Zheng Y. Transcriptional profiling of mouse uterus at pre-implantation stage under VEGF repression. PLoS One 2013; 8:e57287. [PMID: 23468957 PMCID: PMC3585347 DOI: 10.1371/journal.pone.0057287] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 01/18/2013] [Indexed: 11/19/2022] Open
Abstract
Uterus development during pre-implantation stage affects implantation process and embryo growth. Aberrant uterus development is associated with many human reproductive diseases. Among the factors regulating uterus development, vascular remodeling promoters are critical for uterus function and fertility. Vascular endothelial growth factor (VEGF), as one of the major members, has been found to be important in endothelial cell growth and blood vessel development, as well as in non-endothelial cells. VEGF mediation in reproduction has been broadly studied, but VEGF-induced transcriptional machinery during implantation window has not been systematically studied. In this study, a genetically repressed VEGF mouse model was used to analyze uterus transcriptome at gestation 2.5 (G2.5) by Solexa/Illumina's digital gene expression (DGE) system. A number of 831 uterus-specific and 2398 VEGF-regulated genes were identified. Gene ontology (GO) analysis indicated that genes actively involved in uterus development were members of collagen biosynthesis, cell proliferation and cell apoptosis. Uterus-specific genes were enriched in activities of phosphatidyl inositol phosphate kinase, histone H3-K36 demethylation and protein acetylation. Among VEGF-regulated genes, up-regulated were associated with RNA polymerase III activity while down-regulated were strongly related with muscle development. Comparable numbers of antisense transcripts were identified. Expression levels of the antisense transcripts were found tightly correlated with their sense expression levels, an indication of possibly non-specific transcripts generated around the active promoters and enhancers. The antisense transcripts with exceptionally high or low expression levels and the antisense transcripts under VEGF regulation were also identified. These transcripts may be important candidates in regulation of uterus development. This study provides a global survey on genes and antisense transcripts regulated by VEGF in the pre-implantation stage. Results will contribute to further study the candidate genes and pathways in regulating implantation process and related diseases.
Collapse
Affiliation(s)
- Yan Ji
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Xiaodan Lu
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Qingping Zhong
- KLAS and School of Mathematics and Statistics, Northeast Normal University, Changchun, China
| | - Peng Liu
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Yao An
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Yuntao Zhang
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Shujie Zhang
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Ruirui Jia
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Isaias G. Tesfamariam
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Abraha G. Kahsay
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Luqing Zhang
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
- * E-mail: (LQZ); (WSZ); (YWZ)
| | - Wensheng Zhu
- KLAS and School of Mathematics and Statistics, Northeast Normal University, Changchun, China
- * E-mail: (LQZ); (WSZ); (YWZ)
| | - Yaowu Zheng
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
- * E-mail: (LQZ); (WSZ); (YWZ)
| |
Collapse
|
49
|
GUO BIN, ZHANG XUEMING, LI SHIJIE, TIAN XUECHAO, WANG SHOUTANG, LI DANGDANG, LIU JUXIONG, YUE ZHANPENG. Differential Expression and Regulation of Angiopoietin-3 in Mouse Uterus during Preimplantation Period. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2012; 318:316-24. [DOI: 10.1002/jez.b.22449] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- BIN GUO
- College of Animal Science and Veterinary Medicine; Jilin University; Changchun; P. R. China
| | - XUE-MING ZHANG
- College of Animal Science and Veterinary Medicine; Jilin University; Changchun; P. R. China
| | - SHI-JIE LI
- College of Life Science; Northeast Agricultural University; Harbin; P. R. China
| | - XUE-CHAO TIAN
- College of Animal Science and Veterinary Medicine; Jilin University; Changchun; P. R. China
| | - SHOU-TANG WANG
- College of Animal Science and Veterinary Medicine; Jilin University; Changchun; P. R. China
| | - DANG-DANG LI
- College of Animal Science and Veterinary Medicine; Jilin University; Changchun; P. R. China
| | - JU-XIONG LIU
- College of Animal Science and Veterinary Medicine; Jilin University; Changchun; P. R. China
| | - ZHAN-PENG YUE
- College of Animal Science and Veterinary Medicine; Jilin University; Changchun; P. R. China
| |
Collapse
|
50
|
Matsumoto H, Fukui E, Yoshizawa M, Sato E, Daikoku T. Differential expression of the motin family in the peri-implantation mouse uterus and their hormonal regulation. J Reprod Dev 2012; 58:649-53. [PMID: 22813598 DOI: 10.1262/jrd.2012-075] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Increased vascular permeability and angiogenesis are hallmarks of the implantation process in the uterus. Angiomotin (Amot), which is a vascular angiogenesis-related protein, belongs to the motin family. There are two other members of the motin family, angiomotin-like 1 and 2 (Amotl1 and 2), which are also thought to be involved with angiogenesis. In the present study, the distribution of motin mRNAs in the mouse uterus during the peri-implantation period was investigated by in situ hybridization. Amot and Amotl1 were expressed in the stromal cells on days 3 and 4; expressions of Amotl2 during the same period were low. During the postimplantation period, Amot and Amotl1 were expressed in secondary decidual cells, while Amotl2 expression fell to an undetectable level. We also examined hormonal regulation of motin expression by steroid hormone treatment in ovariectomized mice. We found that expression of Amot was induced by P(4) in stromal cells. Additionally, Amotl1 expression was upregulated by both P(4) and estrogen (E(2)) in stromal cells, whereas E(2) increased this gene expression for only a limited time; after 12 h, expression dissipated. In contrast, P(4) regulated the expression of Amotl2 in stromal cells, while E(2) regulated its expression in luminal epithelium cells. Our results demonstrated that Amot, Amotl1, and Amotl2 were differentially expressed in uterine cells during the peri-implantation period, and that their expressions were differentially regulated by P(4) and E(2).
Collapse
Affiliation(s)
- Hiromichi Matsumoto
- Laboratory of Animal Breeding and Reproduction, Division of Animal Science, Faculty of Agriculture, Utsunomiya University, Tochigi 321-8505, Japan.
| | | | | | | | | |
Collapse
|