1
|
Mirani P, Murti K, Lestari PM, Liberty IA, Kesty C, Andrina H, Stevanny B. The Role of CXCR2, MMP-2, and MMP-9 in the Pathogenesis of Placenta Accreta: A Molecular Expression Study. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:461. [PMID: 40142271 PMCID: PMC11944166 DOI: 10.3390/medicina61030461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 02/22/2025] [Accepted: 02/27/2025] [Indexed: 03/28/2025]
Abstract
Background and Objectives: The pathogenesis of placenta accreta spectrum disorder (PASD) is influenced by the inflammatory process. Therefore, the examination of biomarkers related to the inflammatory process, namely matrix metalloproteinase (MMP) and CXC motif chemokine receptor 2 (CXCR2), is expected to bring researchers to a bright spot regarding the pathogenesis of PASD. This study analyzes the role of CXCR2, MMP-2, and MMP-9 in the pathogenesis of PASD. Materials and Methods: An observational study with a case-control design was conducted to assess differences in the mean density of CXCR2, MMP-2, and MMP-9 immunostaining in placental and uterine tissue in 17 patients with PASD and 34 patients without PASD at the Department of Obstetrics and Gynecology, Dr. Mohammad Hoesin Hospital Palembang. The expression of CXCR2, MMP-2, and MMP-9 was measured by immunohistochemistry analysis. The data were analyzed using STATA version 15. Results: There were no significant differences in the mean levels of MMP-2 expression in patients with and without PASD. There were significant differences in the expression of placental CXCR2 (p = 0.003), uterine CXCR2 (p < 0.001), and uterine MMP-9 (p = 0.018) in patients with and without PASD. Conclusions: CXCR2 and MMP-9 may play a role in the pathogenesis of PASD.
Collapse
Affiliation(s)
- Putri Mirani
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Dr. Mohammad Hoesin General Hospital, Faculty of Medicine, Universitas Sriwijaya, Palembang 30114, South Sumatra, Indonesia; (P.M.); (P.M.L.)
| | - Krisna Murti
- Department of Anatomic Pathology, Dr. Mohammad Hoesin General Hospital, Faculty of Medicine, Universitas Sriwijaya, Palembang 30114, South Sumatra, Indonesia
| | - Peby Maulina Lestari
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Dr. Mohammad Hoesin General Hospital, Faculty of Medicine, Universitas Sriwijaya, Palembang 30114, South Sumatra, Indonesia; (P.M.); (P.M.L.)
| | - Iche Andriyani Liberty
- Department of Public Health and Community Medicine, Faculty of Medicine, Universitas Sriwijaya, Palembang 30114, South Sumatra, Indonesia;
| | - Cindy Kesty
- Department of Obstetrics and Gynecology, Dr. Mohammad Hoesin General Hospital, Faculty of Medicine, Universitas Sriwijaya, Palembang 30114, South Sumatra, Indonesia (H.A.); (B.S.)
- World Health Organization, Tropical Diseases Research, Clinical Research Leadership Fellow, Infectious Diseases Data Observatory, University of Oxford, Oxford OX3 7LF, UK
- National Task Force of Reproductive Tract Infection, Indonesian Society of Obstetrics and Gynecology, Jakarta 10320, Indonesia
| | - Hana Andrina
- Department of Obstetrics and Gynecology, Dr. Mohammad Hoesin General Hospital, Faculty of Medicine, Universitas Sriwijaya, Palembang 30114, South Sumatra, Indonesia (H.A.); (B.S.)
| | - Bella Stevanny
- Department of Obstetrics and Gynecology, Dr. Mohammad Hoesin General Hospital, Faculty of Medicine, Universitas Sriwijaya, Palembang 30114, South Sumatra, Indonesia (H.A.); (B.S.)
| |
Collapse
|
2
|
Lin Z, Wu S, Jiang Y, Chen Z, Huang X, Wen Z, Yuan Y. Unraveling the molecular mechanisms driving enhanced invasion capability of extravillous trophoblast cells: a comprehensive review. J Assist Reprod Genet 2024; 41:591-608. [PMID: 38315418 PMCID: PMC10957806 DOI: 10.1007/s10815-024-03036-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/15/2024] [Indexed: 02/07/2024] Open
Abstract
Precise extravillous trophoblast (EVT) invasion is crucial for successful placentation and pregnancy. This review focuses on elucidating the mechanisms that promote heightened EVT invasion. We comprehensively summarize the pivotal roles of hormones, angiogenesis, hypoxia, stress, the extracellular matrix microenvironment, epithelial-to-mesenchymal transition (EMT), immunity, inflammation, programmed cell death, epigenetic modifications, and microbiota in facilitating EVT invasion. The molecular mechanisms underlying enhanced EVT invasion may provide valuable insights into potential pathogenic mechanisms associated with diseases characterized by excessive invasion, such as the placenta accreta spectrum (PAS), thereby offering novel perspectives for managing pregnancy complications related to deficient EVT invasion.
Collapse
Affiliation(s)
- Zihan Lin
- School of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Shuang Wu
- School of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Yinghui Jiang
- School of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Ziqi Chen
- School of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Xiaoye Huang
- School of Pediatrics, Guangzhou Medical University, Guangzhou, China
| | - Zhuofeng Wen
- The Sixth Clinical School of Guangzhou Medical University, Guangzhou, China
| | - Yi Yuan
- School of Pediatrics, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
3
|
Rezaei M, Ghasemi M, Saravani M, Ghahghayi F, Shahraki-Ghadim H, Salimi S. The possible effects of the MTOR polymorphisms on preeclampsia susceptibility, severity, and onset: a case-control study and in silico analysis. Mol Biol Rep 2024; 51:335. [PMID: 38393518 DOI: 10.1007/s11033-023-09190-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 12/21/2023] [Indexed: 02/25/2024]
Abstract
BACKGROUND Preeclampsia (PE) is a gestational complication with developed hypertension and proteinuria. Evidence showed the role of mTOR in various cellular processes. Therefore, this study aimed to evaluate the effects of MTOR polymorphisms on susceptibility, severity, and onset of Preeclampsia (PE). METHODS AND RESULTS A total of 250 PE pregnant women and 258 age-matched control subjects were recruited in this study. To genotype MTOR polymorphisms, the PCR-RFLP method was used. The SpliceAid 2 and PROMO tools were used for in silico analysis. The maternal MTOR rs17036508T/C polymorphism was associated with PE risk in various genetic models. There was no relationship between rs2536T/C and rs2295080T/G polymorphisms and PE. The TTC and TGC haplotypes of rs2536/ rs2295080/ rs17036508 polymorphisms were significantly higher in PE women. Subgroup analysis revealed the association between the MTOR rs2295080 variant and an increased risk of Early-onset PE (EOPE). However, the MTOR rs17036508 was associated with a higher risk of EOPE and Late- Onset PE. In addition, the MTOR rs2295080 could increase the risk of severe PE. The results of the in silico analysis showed that rs17036508 disrupted several binding motifs in the mutant sequence. The PROMO database revealed that the T to C substitution leads to the loss of the TFII-I binding site in the mutant allele. CONCLUSION The MTOR rs17036508T/C polymorphism was associated with PE risk. There was an association between the MTOR rs2295080 variant and an increased risk of EOPE. The MTOR rs17036508T/C and rs2295080T/C variants could disrupt several binding motifs and TFII-I binding respectively.
Collapse
Affiliation(s)
- Mahnaz Rezaei
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Marzieh Ghasemi
- Department of Obstetrics and Gynecology, Pregnancy Health Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
- Pregnancy Health Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mohsen Saravani
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Fatemeh Ghahghayi
- Department of Obstetrics and Gynecology, Pregnancy Health Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Hossein Shahraki-Ghadim
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Saeedeh Salimi
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran.
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Margiana R. Mesenchymal stem cell-derived exosomes in preeclampsia: A next-generation therapeutic tool. Cell Biochem Funct 2024; 42:e3908. [PMID: 38269498 DOI: 10.1002/cbf.3908] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/29/2023] [Accepted: 12/10/2023] [Indexed: 01/26/2024]
Abstract
Preeclampsia (PE) is a major gestational disorder that causes both long- and short-term damage to both the mother and the fetus. Endometrium decidualization and the formation of the placenta are orchestrated by mesenchymal stem cells (MSCs). MSCs obtained from patients with PE exhibit an elevated rate of aging and apoptosis, which impairs the interplay between MSCs and endothelium, trophoblast, and immune cells in the placenta, accelerating the onset of PE. Preclinical and clinical evidence imply that the MSC-based therapy approach for PE is prospective. Importantly, as a novel cell-free approach, MSC-derived exosomes can improve symptoms and maternal-fetal survival in PE models by raising cell metabolism, encouraging angiogenesis balance, and regulating immune responses. Even following allogeneic administration, the likelihood of immune rejection is very limited as a result of the small quantity of exosome membrane-bound proteins. Furthermore, because exosomes do not expand, developing tumors is not probable. As a result, MSC-derived exosomes show superiority over MSCs in terms of safety. For the first time, we outline the properties of MSC-exosomes and highlight their functions and potential as a new paradigm for PE therapy in this review.
Collapse
Affiliation(s)
- Ria Margiana
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Master's Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Andrology Program, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| |
Collapse
|
5
|
Ullah A, Zhao J, Singla RK, Shen B. Pathophysiological impact of CXC and CX3CL1 chemokines in preeclampsia and gestational diabetes mellitus. Front Cell Dev Biol 2023; 11:1272536. [PMID: 37928902 PMCID: PMC10620730 DOI: 10.3389/fcell.2023.1272536] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023] Open
Abstract
Diabetes-related pathophysiological alterations and various female reproductive difficulties were common in pregnant women with gestational diabetes mellitus (GDM), who had 21.1 million live births. Preeclampsia (PE), which increases maternal and fetal morbidity and mortality, affects approximately 3%-5% of pregnancies worldwide. Nevertheless, it is unclear what triggers PE and GDM to develop. Therefore, the development of novel moderator therapy approaches is a crucial advancement. Chemokines regulate physiological defenses and maternal-fetal interaction during healthy and disturbed pregnancies. Chemokines regulate immunity, stem cell trafficking, anti-angiogenesis, and cell attraction. CXC chemokines are usually inflammatory and contribute to numerous reproductive disorders. Fractalkine (CX3CL1) may be membrane-bound or soluble. CX3CL1 aids cell survival during homeostasis and inflammation. Evidence reveals that CXC and CX3CL1 chemokines and their receptors have been the focus of therapeutic discoveries for clinical intervention due to their considerable participation in numerous biological processes. This review aims to give an overview of the functions of CXC and CX3CL1 chemokines and their receptors in the pathophysiology of PE and GDM. Finally, we examined stimulus specificity for CXC and CX3CL1 chemokine expression and synthesis in PE and GDM and preclinical and clinical trials of CXC-based PE and GDM therapies.
Collapse
Affiliation(s)
- Amin Ullah
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine, Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Zhao
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine, Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Rajeev K. Singla
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine, Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Bairong Shen
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine, Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Sui X, Zhang L, Zhang XF, Zhang Y. TRIB3-Regulated Akt Signal Pathway Affects Trophoblast Invasion in the Development of Preeclampsia. Am J Perinatol 2023; 40:1359-1366. [PMID: 34553361 DOI: 10.1055/s-0041-1735872] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVE The aim of the study is to explore the mechanism of tribbles pseudokinase 3 (TRIB3)-regulated Akt pathway in the development of preeclampsia (PE). STUDY DESIGN TRIB3 expression in the placenta of PE patient was determined by quantitative reverse transcriptase polymerase chain reaction and western blotting. Then HTR-8/SVneo or JEG-3 cells were transfected and divided into Mock, Control siRNA, TRIB3 siRNA-1, and TRIB3 siRNA-2 groups. Cell proliferation, invasion, and migration were determined by MTT assay, Transwell assay, and wound healing test, while the expression of TRIB3 and Akt pathway was measured by western blotting. PE rats were treated with TRIB3 siRNA, and blood pressure, 24-hour urinary protein, as well as serum levels of sFlt-1 and vascular endothelial growth factor (VEGF) were measured. RESULTS The placenta of PE patients presented with increased TRIB3 expression. In comparison with Mock group, the proliferation, invasion, and migration of HTR-8/SVneo and JEG-3 cells in TRIB3 siRNA-1 group and TRIB3 siRNA-2 group increased, with decreased TRIB3 expression but enhanced expression of p-Akt/Akt, MMP-2, and MMP-9. Rats in PE group showed increases in mean arterial pressure, SBP, 24-hour urinary protein, and serum sFlt-1 levels, but decreases in serum VEGF levels, fetal weight, and placental efficiency. Moreover, TRIB3 expression was upregulated, while p-Akt/Akt was downregulated in the placenta of rats in PE group. However, indicators above were significantly improved in rats treated with TRIB3 siRNA. CONCLUSION TRIB3 was upregulated in the PE placenta, while silencing TRIB3 activated the Akt signaling pathway to promote the invasion and migration of trophoblast both in vitro and in vivo and ameliorated the development of PE symptoms in the PE rat model. KEY POINTS · The TRIB3 expression was increased in the placenta of PE patient. · Silencing TRIB3 activates Akt signal pathway.. · Silencing TRIB3 improves the pathological process of preeclampsia rat..
Collapse
Affiliation(s)
- Xin Sui
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei Province, China
| | - Lei Zhang
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei Province, China
| | - Xu-Feng Zhang
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei Province, China
| | - Ya Zhang
- Department of Obstetrics and Gynaecology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei Province, China
| |
Collapse
|
7
|
Shi H, Kong R, Miao X, Gou L, Yin X, Ding Y, Cao X, Meng Q, Gu M, Suo F. Decreased PPP1R3G in pre-eclampsia impairs human trophoblast invasion and migration via Akt/MMP-9 signaling pathway. Exp Biol Med (Maywood) 2023; 248:1373-1382. [PMID: 37642261 PMCID: PMC10657594 DOI: 10.1177/15353702231182214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/28/2023] [Indexed: 08/31/2023] Open
Abstract
Pre-eclampsia (PE) is a severe pregnancy complication characterized by impaired trophoblast invasion and spiral artery remodeling and can have serious consequences for both mother and child. Protein phosphatase 1 regulatory subunit 3G (PPP1R3G) is involved in numerous tumor-related biological processes. However, the biological action and underlying mechanisms of PPP1R3G in PE progression remain unclear. We used western blotting and immunohistochemistry to investigate PPP1R3G expression in gestational age-matched pre-eclamptic and normal placental tissues. After lentivirus transfection, wound-healing, Transwell, cell-counting kit-8 (CCK-8), 5-ethynyl-2'-deoxyuridine (EdU), and TdT mediateddUTP Nick End Labeling (TUNEL) assays were used to assess trophoblast migration, invasion, proliferation, and apoptosis, respectively. The relative expression levels of PPP1R3G and the proteins involved in the Akt signaling pathway were determined using western blotting. The results showed that PPP1R3G levels were significantly lower in the placental tissues and GSE74341 microarray of the PE group than those of the healthy control group. We also found that neonatal weight and Apgar score were lower at birth, and peak systolic blood pressure and diastolic blood pressure were higher in the PE group than in the non-PE group. In addition, PPP1R3G knockdown decreased p-Akt/Akt expression and inhibited migration, invasion, and proliferation in HTR-8/SVneo trophoblasts but had no discernible effect on cell apoptosis. Furthermore, PPP1R3G positively regulated matrix metallopeptidase 9 (MMP-9), which was downregulated in placental tissues of pregnant women with PE. These results provided the first evidence that the reduced levels of PPP1R3G might contribute to PE by suppressing the invasion and migration of trophoblasts and targeting the Akt/MMP-9 signaling pathway.
Collapse
Affiliation(s)
- Huimin Shi
- Department of Obstetrics, Xuzhou Cancer Hospital, Xuzhou 221005, Jiangsu Province, China
| | - Renyu Kong
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou 221004, China
| | - Xu Miao
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou 221004, China
| | - Lingshan Gou
- Center for Genetic Medicine, Maternity and Child Health Care Hospital Affiliated to Xuzhou Medical University, 46 Heping Road, Xuzhou 221009, Jiangsu Province, China
| | - Xin Yin
- Center for Genetic Medicine, Maternity and Child Health Care Hospital Affiliated to Xuzhou Medical University, 46 Heping Road, Xuzhou 221009, Jiangsu Province, China
| | - Yuning Ding
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou 221004, China
| | - Xiliang Cao
- Department of Urology, Xuzhou No. 1 People’s Hospital, the Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Qingyong Meng
- Department of Obstetrics, Xuzhou Maternal and Child Health Hospital Affiliated to Xuzhou Medical University, Xuzhou 221009, Jiangsu Province, China
| | - Maosheng Gu
- Center for Genetic Medicine, Maternity and Child Health Care Hospital Affiliated to Xuzhou Medical University, 46 Heping Road, Xuzhou 221009, Jiangsu Province, China
| | - Feng Suo
- Center for Genetic Medicine, Maternity and Child Health Care Hospital Affiliated to Xuzhou Medical University, 46 Heping Road, Xuzhou 221009, Jiangsu Province, China
| |
Collapse
|
8
|
He L, Zhan F, Lu L, Zhang X, Wu J. Role of necroptosis and immune infiltration in preeclampsia: novel insights from bioinformatics analyses. BMC Pregnancy Childbirth 2023; 23:495. [PMID: 37403014 DOI: 10.1186/s12884-023-05821-0] [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: 02/24/2023] [Accepted: 06/29/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND Preeclampsia (PE) is a serious pregnancy complication that can adversely affect the mother and fetus. Necroptosis is a recently discovered new form of programmed cell death involved in the pathological process of various pregnancy complications. Our study aimed to identify the necroptosis-related differentially expressed genes (NRDEGs), create a diagnosis model and related disease subtypes model based on these genes, and further investigate their relationship with immune infiltration. METHODS In this study, we identified NRDEGs by analyzing data from various databases, including Molecular Signatures, GeneCards, and Gene Expression Omnibus (GEO). Using minor absolute shrinkage and selection operator (LASSO) and logistic Cox regression analysis, we developed a novel PE diagnosis model based on NRDEGs. Furthermore, we developed PE subtype models using consensus clustering analysis based on key gene modules screened out by weighted correlation network analysis (WGCNA). Finally, we identified the difference in immune infiltration between the PE and control groups as well as between both PE subtypes by analyzing the immune cell infiltration across combined datasets and PE datasets. RESULTS Our study discovered that the necroptosis pathway was significantly enriched and active in PE samples. We identified nine NRDEGs that involved in this pathway, including BRAF, PAWR, USP22, SYNCRIP, KRT86, MERTK, BAP1, CXCL5, and STK38. Additionally, we developed a diagnostic model based on a regression model including six NRDEGs and identified two PE subtypes: Cluster1 and Cluster2, based on key module genes. Furthermore, correlation analysis showed that the abundance of immune cell infiltration was related to necroptosis genes and PE disease subtypes. CONCLUSION According to the present study, necroptosis is a phenomenon that occurs in PE and is connected to immune cell infiltration. This result suggests that necroptosis and immune-related factors may be the underlying mechanisms of PE pathophysiology. This study opens new avenues for future research into PE's pathogenesis and treatment options.
Collapse
Affiliation(s)
- Lidan He
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Fujian Medical University, Fujian, 350004, China
| | - Feng Zhan
- College of Engineering, Fujian Jiangxia University, Fuzhou, 350108, China
| | - Lin Lu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Fujian Medical University, Fujian, 350004, China
| | - Xia Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Fujian Medical University, Fujian, 350004, China
| | - Jianbo Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Fujian Medical University, Fujian, 350004, China.
| |
Collapse
|
9
|
Alanazi AS, Victor F, Rehman K, Khan YH, Yunusa I, Alzarea AI, Akash MSH, Mallhi TH. Pre-Existing Diabetes Mellitus, Hypertension and KidneyDisease as Risk Factors of Pre-Eclampsia: A Disease of Theories and Its Association with Genetic Polymorphism. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:16690. [PMID: 36554576 PMCID: PMC9778778 DOI: 10.3390/ijerph192416690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 11/25/2022] [Accepted: 12/03/2022] [Indexed: 06/17/2023]
Abstract
Pre-existing diabetes, hypertension and kidney disorders are prominent risk factors of pre-eclampsia (PE). It is a multifactorial pregnancy disorder associated with high blood pressure, proteinuria, and multiorgan failure, which develops after the 20th week of pregnancy. It is one of the most feared pregnancy disorders, as it consumes thousands of fetomaternal lives per annum. According to clinical and pathological studies, the placenta appears to be a key player in the pathogenesis of PE; however, the exact origin of this disorder is still under debate. Defective placentation and angiogenesis are the hallmarks of PE progression. This angiogenic imbalance, together with maternal susceptibility, might determine the severity and clinical presentation of PE. This article comprehensively examines the mechanisms of pathogenesis of PE and current evidence of the factors involved in its progression. Finally, this article will explore the genetic association of PE, various candidate genes, their proposed mechanisms and variants involved in its pathogenesis.
Collapse
Affiliation(s)
- Abdullah Salah Alanazi
- Department of Clinical Pharmacy, College of Pharmacy, Jouf University, Sakaka 72388, Saudi Arabia
| | - Francis Victor
- Department of Pharmacy, University of Chenab, Gujrat 50700, Pakistan
| | - Kanwal Rehman
- Department of Pharmacy, The Women University, Multan 66000, Pakistan
| | - Yusra Habib Khan
- Department of Clinical Pharmacy, College of Pharmacy, Jouf University, Sakaka 72388, Saudi Arabia
| | - Ismaeel Yunusa
- College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA
| | | | | | - Tauqeer Hussain Mallhi
- Department of Clinical Pharmacy, College of Pharmacy, Jouf University, Sakaka 72388, Saudi Arabia
| |
Collapse
|
10
|
Mechanism of Herb Pairs Astragalus mongholicus and Curcuma phaeocaulis Valeton in Treating Gastric Carcinoma: A Network Pharmacology Combines with Differential Analysis and Molecular Docking. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:8361431. [PMID: 35321506 PMCID: PMC8938068 DOI: 10.1155/2022/8361431] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 02/07/2022] [Accepted: 02/11/2022] [Indexed: 02/08/2023]
Abstract
Background Gastric carcinoma (GC) is a kind of digestive tract tumor that is highly malignant and has a very poor prognosis. Although both Astragalus mongholicus (AM, huáng qí) and Curcuma phaeocaulis Valeton (CPV, é zhú) can slow the onset and progression of GC, the mechanism by which AM-CPV works in the treatment of GC is uncertain. Materials and Methods The traditional Chinese medicine network databases TCMSP, TCMID, and ETCM were used to identify the key functional components and associated targets of AM and CPV. To establish a theoretical foundation, the development of gastric cancer (GC) was predicted utilizing a GEO gene chip and TCGA difference analysis mixed with network pharmacology. A herbal-ingredient-target network and a core target-signal pathway network were created using GO and KEGG enrichment analyses. The molecular docking method was used to evaluate seventeen main targets and their compounds. Results Cell activity, reactive oxygen species modification, metabolic regulation, and systemic immune activation may all be involved in the action mechanism of the AM-CPV drug-pair in the treatment of GC. It inhibits the calcium signaling route, the AGE-RAGE signaling system, the cAMP signaling pathway, the PI3K-Akt signaling network, and the MAPK signaling pathway, slowing the progression of GC. The number of inflammatory substances in the tumor microenvironment is reduced, GC cell proliferation is deprived, apoptosis is promoted, and GC progression is retarded through controlling the IL-17 signaling route, TNF signaling pathway, and other inflammation-related pathways. Conclusions The AM-CPV pharmaceutical combination regulates GC treatment via a multitarget, component, and signal pathway with a cooperative and bidirectional regulatory mechanism. Its active constituents may treat GC by regulating the expression of STAT1, MMP9, IL6, HSP90AA1, JUN, CCL2, IFNG, CXCL8, and other targets, as well as activating or inhibiting immune-inflammatory and cancer signaling pathways.
Collapse
|
11
|
Li X, Li C, Wang Y, Cai J, Zhao L, Su Z, Ye H. IGFBP1 inhibits the invasion, migration, and apoptosis of HTR-8/SVneo trophoblast cells in preeclampsia. Hypertens Pregnancy 2022; 41:53-63. [PMID: 35168459 DOI: 10.1080/10641955.2022.2033259] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To investigate the effects and underlying mechanisms of IGFBP1 on the biological functions of trophoblasts in simulated preeclampsia. METHODS IGFBP1 expression in placenta was determined by immunohistochemistry. HTR-8/SVneo cells were stimulated with/without IGFBP1-overexpression and hypoxia-reoxygenation, and the proliferation, invasion, migration, and apoptosis were detected by CCK8, transwell, and flow cytometry, respectively. RESULTS IGFBP1 expression was increased in placenta of preeclampsia. IGFBP1 overexpression inhibited proliferation, invasion, migration, and apoptosis of HTR-8/SVneo cells and induced MMP-26 expression with/without hypoxia-reoxygenation challenge. CONCLUSION IGFBP1 affects biological functions of trophoblasts, and it may play a role in pathophysiology of preeclampsia by inducing MMP-26.
Collapse
Affiliation(s)
- Xiujuan Li
- Department of Clinical Laboratory, Women and Children's Hospital, Xiamen University, School of Medicine, Xiamen, PR China
| | - Chenxi Li
- Department of Clinical Laboratory, Women and Children's Hospital, Xiamen University, School of Medicine, Xiamen, PR China
| | - Ye Wang
- Department of Clinical Laboratory, Women and Children's Hospital, Xiamen University, School of Medicine, Xiamen, PR China
| | - Jianxing Cai
- Department of Clinical Laboratory, Women and Children's Hospital, Xiamen University, School of Medicine, Xiamen, PR China
| | - Li Zhao
- School of Public Health, Xiamen University, Xiamen, PR China
| | - Zhiying Su
- Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, PR China
| | - Huiming Ye
- Department of Clinical Laboratory, Women and Children's Hospital, Xiamen University, School of Medicine, Xiamen, PR China
| |
Collapse
|
12
|
Zhang S, Ding J, Wang J, Yin T, Zhang Y, Yang J. CXCL5 Downregulation in Villous Tissue Is Correlated With Recurrent Spontaneous Abortion. Front Immunol 2021; 12:717483. [PMID: 34603292 PMCID: PMC8486294 DOI: 10.3389/fimmu.2021.717483] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/30/2021] [Indexed: 11/18/2022] Open
Abstract
Recurrent spontaneous abortion (RSA) affects 5% of childbearing-age women worldwide. Inadequate trophoblast invasion is one of the main reasons for the development of RSA; however, the underlying molecular mechanisms for RSA have not been fully understood, and further explanation is urgently needed. C-X-C motif chemokine ligand 5 (CXCL5) is reported to contribute to the invasion of cancer cells, and its aberrant expression is associated with the cellular process of tumor pathology. Considering the high behavioral similarity between trophoblast cells and cancer cells, we hypothesized that CXCL5 may influence trophoblast invasion, and its expression levels in villous tissue may be correlated with RSA. In this study, we firstly investigated the CXCL5 expression in placental villous tissues of 15 RSA patients and 13 control patients, and the results showed that CXCL5 levels were significantly lower in villous tissue from RSA patients than those of the controls. Further in vitro experiments presented that recombinant human CXCL5 can enhance trophoblast migration, invasion, and epithelial-to-mesenchymal transition (EMT) process. We also demonstrated that CXCL5 exerted these effects on trophoblast cells through PI3K/AKT/ERK1/2 signaling pathway. In conclusion, these data indicate that CXCL5 downregulation in human villous tissue is correlated with RSA. Additionally, we found that estrogen, progesterone, human chorionic gonadotropin, and decidual stromal cells can regulate CXCL5 and chemokine receptor 2 (CXCR2) expression of trophoblast in a cell manner.
Collapse
Affiliation(s)
- Sainan Zhang
- Reproductive Medical Center, Renmin Hospital of Wuhan University & Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| | - Jinli Ding
- Reproductive Medical Center, Renmin Hospital of Wuhan University & Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| | - Jiayu Wang
- Reproductive Medical Center, Renmin Hospital of Wuhan University & Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| | - Tailang Yin
- Reproductive Medical Center, Renmin Hospital of Wuhan University & Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| | - Yan Zhang
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jing Yang
- Reproductive Medical Center, Renmin Hospital of Wuhan University & Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| |
Collapse
|
13
|
Zhang X, Li Y, Huang C, Liu S, Chen X, Yu S, Diao L, Zeng Y. The role of decidual natural killer cell derived soluble factors in early pregnancy. Am J Reprod Immunol 2021; 86:e13477. [PMID: 34051025 DOI: 10.1111/aji.13477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/20/2021] [Accepted: 05/22/2021] [Indexed: 11/29/2022] Open
Abstract
Decidual natural killer cell (dNK), the predominant decidual lymphocytes in early pregnancy, are primarily identified based on their CD56bright CD16- phenotype and play an important role in maintaining immune tolerance at the maternal-fetal interface. dNK dysfunction reportedly leads to pathological pregnancy. Indeed, various dNK-derived soluble factors are involved in a series of key processes related to pregnancy outcomes. In this review, we summarize the roles of these dNK-derived factors in immune tolerance and embryonic development to improve the current understanding regarding the physiological and pathological mechanisms that occur during pregnancy, while potentially informing the development of effective therapeutics.
Collapse
Affiliation(s)
- Xueling Zhang
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, 518045, Guangdong, China
| | - Yuye Li
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, 518045, Guangdong, China
| | - Chunyu Huang
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, 518045, Guangdong, China
| | - Su Liu
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, 518045, Guangdong, China
| | - Xian Chen
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, 518045, Guangdong, China
| | - Shuyi Yu
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, 518045, Guangdong, China
| | - Lianghui Diao
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, 518045, Guangdong, China
| | - Yong Zeng
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, 518045, Guangdong, China
| |
Collapse
|
14
|
Investigational Antiviral Therapy Models for the Prevention and Treatment of Congenital Cytomegalovirus Infection during Pregnancy. Antimicrob Agents Chemother 2020; 65:AAC.01627-20. [PMID: 33077661 DOI: 10.1128/aac.01627-20] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/14/2020] [Indexed: 12/29/2022] Open
Abstract
Congenital cytomegalovirus (HCMV) infection may cause significant fetal malformation, lifelong disease, and, in severe cases, fetal or neonatal death. Placental infection with HCMV is the major mechanism of mother-to-child transmission (MTCT) and fetal injury. Thus, any pharmaceutical antiviral interference to reduce viral load may reduce placental damage, MTCT, and fetal disease. However, there is currently no licensed HCMV antiviral for use during pregnancy. In this study, aciclovir and the HCMV-specific antivirals letermovir, maribavir, and cidofovir were compared with ganciclovir for antiviral effects in model systems of pregnancy, including first-trimester TEV-1 trophoblast cell cultures and third-trimester ex vivo placental explant histocultures. HCMV-infected trophoblasts at 7 days postinfection (dpi) showed an EC50 of 21 μM for aciclovir, 0.0007 μM for letermovir, 0.11 μM for maribavir, and 0.29 μM for cidofovir, relative to 0.42 μM for ganciclovir. Antivirals added at 10 μM showed no cytotoxic effects and did not affect trophoblast cell proliferation (P > 0.9999). Multiple-round HCMV replication measured at 7 dpi showed letermovir, maribavir, and cidofovir treatment inhibited immediate early, early, and true late viral protein expression as assayed on Western blots. Antiviral treatment of HCMV-infected placental explants showed significant inhibition (P < 0.05) of viral replication with letermovir (83.3%), maribavir (83.6%), cidofovir (89.3%), and ganciclovir (82.4%), but not aciclovir (P > 0.9999). In ex vivo model systems, recently trialed HCMV antivirals letermovir and maribavir were effective at inhibiting HCMV replication. They partly fulfil requirements for use as safe and effective therapeutics during pregnancy to control congenital HCMV. Clinical trials of these newer agents would assist assessment of their utility in pregnancy.
Collapse
|
15
|
Huang J, Ling Z, Zhong H, Yin Y, Qian Y, Gao M, Ding H, Cheng Q, Jia R. Distinct expression profiles of peptides in placentae from preeclampsia and normal pregnancies. Sci Rep 2020; 10:17558. [PMID: 33067549 PMCID: PMC7567870 DOI: 10.1038/s41598-020-74840-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 10/06/2020] [Indexed: 01/03/2023] Open
Abstract
This study sought to identify potential bioactive peptides from the placenta that are involved in preeclampsia (PE) to obtain information about the prediction, diagnosis and treatment of PE. The liquid chromatography/mass spectrometry was used to perform a comparative analysis of placental peptides in normal and PE pregnancies. Gene ontology (GO), pathway analysis and ingenuity pathway analysis (IPA) were used to evaluate the underlying biological function of the differential peptides based on their protein precursors. Transwell assays and qPCR were used to study the effect of the identified bioactive peptides on the function of HTR-8/SVneo cells. A total of 392 upregulated peptides and 420 downregulated peptides were identified (absolute fold change ≥ 2 and adjusted P value < 0.05). The GO analysis, pathway analysis, and IPA revealed that these differentially expressed peptides play a role in PE. In addition, the up-regulated peptide “DQSATALHFLGRVANPLSTA” derived from Angiotensinogen exhibited effect on the invasiveness of HTR-8/SVneo cells. The current preliminary research not only provides a new research direction for studying the pathogenesis of PE, but also brings new insights for the prediction, diagnosis and treatment of PE.
Collapse
Affiliation(s)
- Jin Huang
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, Jiangsu, China.,Yixing People's Hospital, YiXing, 214200, Jiangsu, China
| | - Zhonghui Ling
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, Jiangsu, China
| | - Hong Zhong
- Fourth Clinical Medicine College, Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Yadong Yin
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, Jiangsu, China
| | - Yating Qian
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, Jiangsu, China
| | - Mingming Gao
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, Jiangsu, China.,Fourth Clinical Medicine College, Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Hongjuan Ding
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, Jiangsu, China
| | - Qing Cheng
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, Jiangsu, China.
| | - Ruizhe Jia
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, Jiangsu, China.
| |
Collapse
|
16
|
Tu C, Tao F, Qin Y, Wu M, Cheng J, Xie M, Shen B, Ren J, Xu X, Huang D, Chen H. Serum proteins differentially expressed in early- and late-onset preeclampsia assessed using iTRAQ proteomics and bioinformatics analyses. PeerJ 2020; 8:e9753. [PMID: 32953262 PMCID: PMC7473043 DOI: 10.7717/peerj.9753] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 07/28/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Preeclampsia remains a serious disorder that puts at risk the lives of perinatal mothers and infants worldwide. This study assessed potential pathogenic mechanisms underlying preeclampsia by investigating differentially expressed proteins (DEPs) in the serum of patients with early-onset preeclampsia (EOPE) and late-onset preeclampsia (LOPE) compared with healthy pregnant women. METHODS Blood samples were collected from four women with EOPE, four women with LOPE, and eight women with normal pregnancies, with four women providing control samples for each preeclampsia group. Serum proteins were identified by isobaric tags for relative and absolute quantitation combined with liquid chromatography-tandem mass spectrometry. Serum proteins with differences in their levels compared with control groups of at least 1.2 fold-changes and that were also statistically significantly different between the groups at P < 0.05 were further analyzed. Bioinformatics analyses, including gene ontology and Kyoto Encyclopedia of Genes and Genomes signaling pathway analyses, were used to determine the key proteins and signaling pathways associated with the development of PE and to determine those DEPs that differed between women with EOPE and those with LOPE. Key protein identified by mass spectrometry was verified by enzyme linked immunosorbent assay (ELISA). RESULTS Compared with serum samples from healthy pregnant women, those from women with EOPE displayed 70 proteins that were differentially expressed with significance. Among them, 51 proteins were significantly upregulated and 19 proteins were significantly downregulated. In serum samples from women with LOPE, 24 DEPs were identified , with 10 proteins significantly upregulated and 14 proteins significantly downregulated compared with healthy pregnant women. Bioinformatics analyses indicated that DEPs in both the EOPE and LOPE groups were associated with abnormalities in the activation of the coagulation cascade and complement system as well as with lipid metabolism. In addition, 19 DEPs in the EOPE group were closely related to placental development or invasion of tumor cells. Downregulationof pregnancy-specific beta-1-glycoprotein 9 (PSG9) in the LOPE group was confirmed by ELISA. CONCLUSION The pathogenesis of EOPE and LOPE appeared to be associated with coagulation cascade activation, lipid metabolism, and complement activation. However, the pathogenesis of EOPE also involved processes associated with greater placental injury. This study provided several new proteins in the serum which may be valuable for clinical diagnosis of EOPE and LOPE, and offered potential mechanisms underpinning the development of these disorders.
Collapse
Affiliation(s)
- Chengcheng Tu
- Department of Obstetrics and Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| | - Feng Tao
- Department of Obstetrics and Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| | - Ying Qin
- School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Mingzhu Wu
- Department of Obstetrics and Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| | - Ji Cheng
- Department of Obstetrics and Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| | - Min Xie
- School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Bing Shen
- School of Basic Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Junjiao Ren
- Department of Science and Education, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| | - Xiaohong Xu
- Department of Clinical Laboratory, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| | - Dayan Huang
- Department of Science and Education, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| | - Hongbo Chen
- Department of Obstetrics and Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
17
|
Jena MK, Sharma NR, Petitt M, Maulik D, Nayak NR. Pathogenesis of Preeclampsia and Therapeutic Approaches Targeting the Placenta. Biomolecules 2020; 10:biom10060953. [PMID: 32599856 PMCID: PMC7357118 DOI: 10.3390/biom10060953] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/20/2020] [Accepted: 06/21/2020] [Indexed: 02/07/2023] Open
Abstract
Preeclampsia (PE) is a serious pregnancy complication, affecting about 5–7% of pregnancies worldwide and is characterized by hypertension and damage to multiple maternal organs, primarily the liver and kidneys. PE usually begins after 20 weeks’ gestation and, if left untreated, can lead to serious complications and lifelong disabilities—even death—in both the mother and the infant. As delivery is the only cure for the disease, treatment is primarily focused on the management of blood pressure and other clinical symptoms. The pathogenesis of PE is still not clear. Abnormal spiral artery remodeling, placental ischemia and a resulting increase in the circulating levels of vascular endothelial growth factor receptor-1 (VEGFR-1), also called soluble fms-like tyrosine kinase-1 (sFlt-1), are believed to be among the primary pathologies associated with PE. sFlt-1 is produced mainly in the placenta during pregnancy and acts as a decoy receptor, binding to free VEGF (VEGF-A) and placental growth factor (PlGF), resulting in the decreased bioavailability of each to target cells. Despite the pathogenic effects of increased sFlt-1 on the maternal vasculature, recent studies from our laboratory and others have strongly indicated that the increase in sFlt-1 in PE may fulfill critical protective functions in preeclamptic pregnancies. Thus, further studies on the roles of sFlt-1 in normal and preeclamptic pregnancies are warranted for the development of therapeutic strategies targeting VEGF signaling for the treatment of PE. Another impediment to the treatment of PE is the lack of suitable methods for delivery of cargo to placental cells, as PE is believed to be of placental origin and most available therapies for PE adversely impact both the mother and the fetus. The present review discusses the pathogenesis of PE, the complex role of sFlt-1 in maternal disease and fetal protection, and the recently developed placenta-targeted drug delivery system for the potential treatment of PE with candidate therapeutic agents.
Collapse
Affiliation(s)
- Manoj Kumar Jena
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University (LPU), Phagwara, Punjab 144411, India;
- Correspondence:
| | - Neeta Raj Sharma
- Department of Biotechnology, School of Bioengineering and Biosciences, Lovely Professional University (LPU), Phagwara, Punjab 144411, India;
| | - Matthew Petitt
- Redwood Biomedical Editing, Redwood City, CA 94061, USA;
| | - Devika Maulik
- Department of Obstetrics and Gynecology, UMKC School of Medicine, Kansas City, MO 64108, USA; (D.M.); (N.R.N.)
| | - Nihar Ranjan Nayak
- Department of Obstetrics and Gynecology, UMKC School of Medicine, Kansas City, MO 64108, USA; (D.M.); (N.R.N.)
| |
Collapse
|
18
|
Circulating Neutrophils Do Not Predict Subclinical Coronary Artery Disease in Women with Former Preeclampsia. Cells 2020; 9:cells9020468. [PMID: 32085575 PMCID: PMC7072843 DOI: 10.3390/cells9020468] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/13/2020] [Accepted: 02/14/2020] [Indexed: 01/30/2023] Open
Abstract
: Introduction: Preeclampsia (PE) represents a hypertensive pregnancy disorder that is associated with increased cardiovascular disease (CVD) risk. This increased risk has been attributed to accelerated atherosclerosis, with inflammation being a major contributor. Neutrophils play an important role in the onset and progression of atherosclerosis and have been associated with vascular damage in the placenta as well as the chronic inflammatory state in women with PE. We therefore investigated whether circulating neutrophil numbers or reactivity were associated with the presence and severity of subclinical atherosclerosis in women with a history of PE. METHODS Women aged 45-60 years with a 10 to 20 years earlier history of early onset preeclampsia (delivery <34 weeks of gestation) (n = 90), but without symptomatic CVD burden were screened for the presence of subclinical coronary artery disease (CAD) using both contrast-enhanced and non-contrast coronary CT angiography. Subclinical CAD was defined as a coronary artery calcium (CAC) score ≥100 Agatston Units and/or ≥50% coronary luminal stenosis. We assessed whether the numbers and activity of circulating neutrophils were associated with the presence of subclinical CAD and as secondary outcome measurements, with the presence of any calcium (CAC score > 0 AU) or stenosis, categorized as absent (0%), minimal to mild (>0 and <50%), and moderate to severe (≥50%) narrowing of the coronary artery. Blood was drawn just before CT and neutrophil numbers were assessed by flow cytometry. In addition, the presence of the chemokine receptors CXCR2 and CXCR4, which are known to be instrumental in neutrophil recruitment, and neutrophil activity upon stimulation with the bacterial peptide N-Formylmethionyl-leucyl-phenylalanine (fMLF) was assessed by flow cytometry. RESULTS Of the participating women, with an average age of 49 years, 13% (12 out of 90) presented with subclinical signs of CAD (CAC score ≥100 AU and/or ≥50% luminal stenosis), and 37% (33 out of 90) had a positive CAC score (>0). Total white blood cell count and neutrophil counts were not associated with the presence of subclinical CAD or with a positive CAC score. When assessing the presence of the chemokine receptors CXCR4 and CXCR2, we observed a slight decrease of neutrophil CXCR2 expression in women with CAC (median MFI 22.0 [interquartile range (IQR) 20.2-23.8]) compared to women without CAC (23.8 [IQR 21.6-25.6], p = 0.02). We observed no differences regarding neutrophil CXCR4 expression. In addition, expression of the early activity marker CD35 was slightly lower on neutrophils of women with subclinical CAD (median MFI 1.6 [IQR 1.5-1.9] compared to 1.9 [IQR 1.7-2.1] in women without CAD, p = 0.02). However, for all findings, statistical significance disappeared after adjustment for multiple testing. CONCLUSION Our findings indicate that neutrophil counts and (re)activity are not directly associated with silent CAD disease burden and as such are not suitable as biomarkers to predict the presence of subclinical CAD in a high-risk population of women with a history of preeclampsia.
Collapse
|
19
|
Timaxian C, Raymond-Letron I, Bouclier C, Gulliver L, Le Corre L, Chébli K, Guillou A, Mollard P, Balabanian K, Lazennec G. The health status alters the pituitary function and reproduction of mice in a Cxcr2-dependent manner. Life Sci Alliance 2020; 3:3/3/e201900599. [PMID: 32041848 PMCID: PMC7010316 DOI: 10.26508/lsa.201900599] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/28/2020] [Accepted: 01/29/2020] [Indexed: 12/18/2022] Open
Abstract
This study explores the effects of microbiota on reproductive function of Cxcr2 knockout animals. Cxcr2 is involved in the control of pituitary action and the subsequent development of mammary gland, uterus and ovary. Microbiota and chronic infections can affect not only immune status, but also the overall physiology of animals. Here, we report that chronic infections dramatically modify the phenotype of Cxcr2 KO mice, impairing in particular, their reproduction ability. We show that exposure of Cxcr2 KO females to multiple types of chronic infections prevents their ability to cycle, reduces the development of the mammary gland and alters the morphology of the uterus due to an impairment of ovary function. Mammary gland and ovary transplantation demonstrated that the hormonal contexture was playing a crucial role in this phenomenon. This was further evidenced by alterations to circulating levels of sex steroid and pituitary hormones. By analyzing at the molecular level the mechanisms of pituitary dysfunction, we showed that in the absence of Cxcr2, bystander infections affect leukocyte migration, adhesion, and function, as well as ion transport, synaptic function behavior, and reproduction pathways. Taken together, these data reveal that a chemokine receptor plays a direct role in pituitary function and reproduction in the context of chronic infections.
Collapse
Affiliation(s)
- Colin Timaxian
- Centre National de la Recherche Scientifique (CNRS), SYS2DIAG-ALCEDIAG, Cap Delta, Montpellier, France.,CNRS, Groupement de Recherche 3697 "Microenvironment of Tumor Niches," Micronit, France
| | - Isabelle Raymond-Letron
- Department of Histopathology, National Veterinary School of Toulouse, France and Platform of Experimental and Compared Histopathology, STROMALab, Unité de recherche mixte (UMR) Université Paul Sabatier/CNRS 5223, Etablissement français du sang, Institut national de la santé et de la recherche médicale (Inserm) U1031, Toulouse, France
| | - Céline Bouclier
- Centre National de la Recherche Scientifique (CNRS), SYS2DIAG-ALCEDIAG, Cap Delta, Montpellier, France
| | | | - Ludovic Le Corre
- Nutrition et Toxicologie Alimentaire (NUTOX) Laboratory - INSERM Lipides, Nutrition, Cancer UMR 1231 - AgrosupDijon, Dijon, France
| | - Karim Chébli
- Equipe Metazoan Messenger RNAs Metabolism, Montpellier, France
| | - Anne Guillou
- Institut de Génomique Fonctionnelle, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Patrice Mollard
- Institut de Génomique Fonctionnelle, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Karl Balabanian
- CNRS, Groupement de Recherche 3697 "Microenvironment of Tumor Niches," Micronit, France.,Université de Paris, Institut de Recherche Saint-Louis, EMiLy, INSERM U1160, Paris, France
| | - Gwendal Lazennec
- Centre National de la Recherche Scientifique (CNRS), SYS2DIAG-ALCEDIAG, Cap Delta, Montpellier, France .,CNRS, Groupement de Recherche 3697 "Microenvironment of Tumor Niches," Micronit, France
| |
Collapse
|
20
|
Chen Y, Ding H, Wei M, Zha W, Guan S, Liu N, Li Y, Tan Y, Wang Y, Wu F. MSC-Secreted Exosomal H19 Promotes Trophoblast Cell Invasion and Migration by Downregulating let-7b and Upregulating FOXO1. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 19:1237-1249. [PMID: 32069774 PMCID: PMC7026285 DOI: 10.1016/j.omtn.2019.11.031] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 11/13/2019] [Accepted: 11/24/2019] [Indexed: 12/12/2022]
Abstract
Exosomes perform important functions for intercellular communication through extracellular signaling pathways, leading to the regulation of important biological processes, including cell proliferation, but also systemic dysfunctions such as preeclampsia (PE). However, the inhibitory effects of mesenchymal stem cell (MSCs)-derived exosomes in PE remain largely unknown. Thus, we assessed the possibility that exosomes could transport long non-coding RNA H19 and the correlation between H19 and the apoptosis of trophoblast cells. The expression of microRNA let-7b and forkhead box protein O1 (FOXO1) was characterized in placental tissues of PE patients. Gain- and loss-of-function experiments were performed to examine the roles of FOXO1 and let-7b in trophoblast cells. Interactions between let-7b and H19 as well as between let-7b and FOXO1 were confirmed by a dual-luciferase reporter assay, RNA pull-down, and RNA immunoprecipitation. HTR-8/SVneo cells were co-cultured with exosomes derived from MSCs overexpressing H19, followed by invasion, migration, and apoptosis assessments of trophoblast cells. We found that let-7b was highly expressed and FOXO1 was poorly expressed in placental tissues of PE patients. Furthermore, H19 acts as a competitive endogenous RNA against let-7b, and let-7b directly targeted FOXO1. Moreover, H19 could be transferred to trophoblast cells via MSC-secreted exosomes. MSC-derived exosomes overexpressing H19 decreased let-7b, increased FOXO1, and activated the protein kinase B (AKT) signaling pathway, thus increasing invasion and migration and inhibiting apoptosis of trophoblast cells. These results suggest that MSC-derived exosomes overexpressing H19 may be a novel direction for therapeutic strategies against PE.
Collapse
Affiliation(s)
- Yang Chen
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Haiyan Ding
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Min Wei
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Wenhui Zha
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Shuang Guan
- Department of Rehabilitation, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Ning Liu
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Yang Li
- Center of Reproductive Medicine, Center of Prenatal Diagnosis, The First Hospital of JiLin University, Changchun 130041, P.R. China
| | - Yuan Tan
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Yan Wang
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Fuju Wu
- Department of Gynecology and Obstetrics, The Second Hospital of Jilin University, Changchun 130041, P.R. China.
| |
Collapse
|
21
|
Jia X, Lu M, Rui C, Xiao Y. Consensus-Expressed CXCL8 and MMP9 Identified by Meta-Analyzed Perineural Invasion Gene Signature in Gastric Cancer Microarray Data. Front Genet 2019; 10:851. [PMID: 31681401 PMCID: PMC6798046 DOI: 10.3389/fgene.2019.00851] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 08/15/2019] [Indexed: 12/21/2022] Open
Abstract
As an underrecognized route of cancer metastasis, perineural invasion (PNI) is defined as the neoplastic invasion of nerves, which can be targeted to inhibit the metastasis of malignant cancer. However, the mechanism underlying PNI in cancer is largely unknown. We constructed a PNI gene signature based on a Pathway Studio–mediated literature screen and investigated the relevant genes in a gastric cancer model. Thus, a total of 467 studies/datasets were retrieved from the Gene Expression Omnibus database using the keyword “gastric cancer,” among which 13 studies that focused on gene expression profiling were further manually inspected and selected. Furthermore, the constructed PNI gene signature (104 genes) expression was meta-analyzed, and the consensus-expressed C-X-C motif chemokine ligand 8 (CXCL8) and matrix metallopeptidase 9 (MMP9) (p < 0.01, |log fold change| >1) were detected. Importantly, the disease-free survival was significantly worse in patients with high expressions of CXCL8 and MMP9 than in those with low expressions (p = 0.05). Moreover, multiple linear regression analysis showed that the population region (country) was associated with the expressions of both CXCL8 and MMP9. In conclusion, these data suggest that the coexpression of CXCL8 and MMP9 could be an early detection marker for PNI, with a potential to be utilized as individual therapy targets for early treatment to prevent PNI-related cancer metastasis.
Collapse
Affiliation(s)
- Xiuzhi Jia
- Central Lab of Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Minjia Lu
- Research Department, Hangzhou Beiwo Meditech Co., Ltd, Hangzhou, Zhejiang Province, China
| | - Chen Rui
- Central Lab of Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Ying Xiao
- Central Lab of Biomedical Research Center, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
22
|
Li L, Huang X, He Z, Xiong Y, Fang Q. miRNA-210-3p regulates trophoblast proliferation and invasiveness through fibroblast growth factor 1 in selective intrauterine growth restriction. J Cell Mol Med 2019; 23:4422-4433. [PMID: 30993882 PMCID: PMC6533475 DOI: 10.1111/jcmm.14335] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/15/2019] [Accepted: 03/30/2019] [Indexed: 12/25/2022] Open
Abstract
Selective intrauterine growth restriction (sIUGR), which affects approximately 10%-15% of monochorionic (MC) twin pregnancies, is highly associated with intrauterine foetal death and neurological impairment in both twins. Data suggest that unequal sharing of the single placenta is the main contributor to birth weight discordance. While MC twins and their placenta derive from a single zygote and harbour almost identical genetic material, the underlying mechanisms of phenotypic discrepancies in MC twins remain unclear. MicroRNAs are small non-coding RNA molecules that regulate gene expression but do not change the DNA sequence. Our preliminary study showed that microRNA-210-3p (miR-210-3p) was significantly upregulated in the placental share of the smaller sIUGR twin. Here, we investigate the potential role of miR-210-3p in placental dysplasia, which generally results from dysfunction of trophoblast cells. Functional analysis revealed that miR-210-3p, induced by hypoxia-inducible factor 1α (HIF1α) under hypoxic conditions, suppressed the proliferation and invasiveness of trophoblast cell lines. Further RNA sequencing analysis and luciferase reporter assays were performed, revealing that fibroblast growth factor 1 (FGF1) is an influential target gene of miR-210-3p. Moreover, correlations among miR-210-3p levels, HIF1α and FGF1 expression and the smaller placental share were validated in sIUGR specimens. These findings suggest that upregulation of miR-210-3p may contribute to impaired placentation of the smaller twin by decreasing FGF1 expression in sIUGR.
Collapse
Affiliation(s)
- Lin Li
- Department of Obstetrics and Gynecology, Fetal Medicine CenterThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Xuan Huang
- Department of Obstetrics and Gynecology, Fetal Medicine CenterThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Zhiming He
- Department of Obstetrics and Gynecology, Fetal Medicine CenterThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Yuanyan Xiong
- Key Laboratory of Gene Engineering of the Ministry of Education and State Key Laboratory of Biocontrol, School of Life SciencesSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Qun Fang
- Department of Obstetrics and Gynecology, Fetal Medicine CenterThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdongChina
| |
Collapse
|
23
|
Hypoxia-induced microRNA-141 regulates trophoblast apoptosis, invasion, and vascularization by blocking CXCL12β/CXCR2/4 signal transduction. Biomed Pharmacother 2019; 116:108836. [PMID: 31004838 DOI: 10.1016/j.biopha.2019.108836] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 03/19/2019] [Accepted: 03/29/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND An impaired trophoblast invasion ability contributes to the development of pre-eclampsia (PE), and can be induced by the altered expression of various microRNAs (miRs). MiR-141 and CXCL12β (C-X-C motif chemokine ligand 12) signaling regulate trophoblast invasion and vascularization capabilities during PE pathogenesis; however, their interactions and underlying mechanisms of action remain unclear. We investigated how miR-141 modulates trophoblast invasion, with a focus on its interaction with CXCL12β signaling. METHODS A PE model was established by using HTR-8/SVneo cells, which were first cultured with 2% O2 for 48 h, and then with 5% O2. The expression of miR-141 in human villous trophoblast HTR-8/SVneo cells was modulated with mimics or an inhibitor, and analyzed by quantitative RT-PCR. CXCL12β levels were determined by ELISA. Cell apoptosis was determined by flow cytometry, and the invasion and vascularization capabilities of trophoblasts were evaluated by Transwell and tube formation assays, respectively. Binding of miR-141 with CXCL12β mRNA was verified by the dual luciferase assay. Protein levels were estimated by western blotting. RESULTS MiR-141 expression was significantly induced by hypoxia in HTR-8/SVneo cells. MiR-141 was found to promote apoptosis and inhibit the invasion and vascularization abilities of HTR-8/SVneo cells under conditions of hypoxia. MiR-141 could directly bind with the 3'UTR region of CXCL12β mRNA and inhibit its translation. In addition, we proved that miR-141 could inhibit the invasion and vascularization abilities, and promote the apoptosis of HTR-8/SVneo cells by targeting CXCL12β under hypoxic conditions. Furthermore, we demonstrated that arachidonic acid could reverse the invasion and apoptosis abilities of HTR-8/SVneo cells mediated by CXCL12β during hypoxia. In terms of mechanism, MiR-141 could downregulate MMP2, p62, and LC3B expression, and upregulate ROCK1 and RhoA expression in HTR-8/SVneo cells by targeting the CXCL12β gene during hypoxia. The effects of CXCL12βon HTR-8/SVneo cells could be reversed by arachidonic acid (ARA). CONCLUSION Induction of miR-141 by hypoxia promotes apoptosis, and inhibits the invasion and vascularization capabilities of HTR-8/SVneo cells by suppressing the CXCL12β and CXCR2/4 signaling pathways.
Collapse
|
24
|
Lai WS, Ding YL. GNG7 silencing promotes the proliferation and differentiation of placental cytotrophoblasts in preeclampsia rats through activation of the mTOR signaling pathway. Int J Mol Med 2019; 43:1939-1950. [PMID: 30864685 PMCID: PMC6443336 DOI: 10.3892/ijmm.2019.4129] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 02/08/2019] [Indexed: 12/25/2022] Open
Abstract
Preeclampsia (PE) is a pathological condition that manifests during pregnancy as the occurrence of an abnormal urine protein level and increased blood pressure due to inadequate cytotrophoblast invasion. To elucidate the mechanism underlying PE, the present study primarily focused on the regulatory effects and mechanism of the G protein γ 7 (GNG7) on placental cytotrophoblasts in a rat PE model. Initially, the PE model was established with 45 specific pathogen‑free Sprague‑Dawley rats (30 females and 15 males). The expression patterns of GNG7, 4E‑binding protein 1 (4E‑BP1), phosphoprotein 70 ribosomal protein S6 kinase (p70S6K) and mammalian target of rapamycin (mTOR) were examined in the PE rats. Placental cytotrophoblasts isolated from normal and PE rats were treated with a small interfering RNA against GNG7, mTOR signaling pathway activator (HIV‑1 Tat) or inhibitor (rapamycin). Following treatment, cell proliferation, differentiation and apoptosis were evaluated, and mTOR signaling pathway‑related factors (4E‑BP1, p70S6K and mTOR), cell proliferation‑related factors (vascular endothelial growth factor and transforming growth factor‑β1), differentiation‑related factors [activator protein‑2 (AP‑2)α and AP‑2γ], and apoptosis‑related factors [B‑cell lymphoma 2 (Bcl‑2) and Bcl‑2‑associated X protein] were determined. Finally, soluble fms‑like tyrosine kinase 1 (sFlt‑1) and soluble endoglin (sEng) levels were measured via enzyme‑linked immunosorbent assay. Initially, the mTOR signaling pathway was inactivated in the placental tissues and cytotrophoblasts in the PE rats. Silencing GNG7 reduced the levels of sFlt‑1 and sEng and activated the mTOR signaling pathway. Silencing of GNG7 or activation of the mTOR signaling pathway enhanced cell proliferation and differentiation, but inhibited the apoptosis of placental cytotrophoblasts in the PE rats. Taken together, the results showed that GNG7 silencing repressed apoptosis and enhanced the proliferation and differentiation of placental cytotrophoblasts in PE rats through activation of the mTOR signaling pathway.
Collapse
Affiliation(s)
- Wei-Si Lai
- Department of Obstetrics and Gynecology, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Yi-Ling Ding
- Department of Obstetrics and Gynecology, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
25
|
Schiebinger G, Shu J, Tabaka M, Cleary B, Subramanian V, Solomon A, Gould J, Liu S, Lin S, Berube P, Lee L, Chen J, Brumbaugh J, Rigollet P, Hochedlinger K, Jaenisch R, Regev A, Lander ES. Optimal-Transport Analysis of Single-Cell Gene Expression Identifies Developmental Trajectories in Reprogramming. Cell 2019; 176:928-943.e22. [PMID: 30712874 PMCID: PMC6402800 DOI: 10.1016/j.cell.2019.01.006] [Citation(s) in RCA: 330] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/15/2018] [Accepted: 01/02/2019] [Indexed: 12/18/2022]
Abstract
Understanding the molecular programs that guide differentiation during development is a major challenge. Here, we introduce Waddington-OT, an approach for studying developmental time courses to infer ancestor-descendant fates and model the regulatory programs that underlie them. We apply the method to reconstruct the landscape of reprogramming from 315,000 single-cell RNA sequencing (scRNA-seq) profiles, collected at half-day intervals across 18 days. The results reveal a wider range of developmental programs than previously characterized. Cells gradually adopt either a terminal stromal state or a mesenchymal-to-epithelial transition state. The latter gives rise to populations related to pluripotent, extra-embryonic, and neural cells, with each harboring multiple finer subpopulations. The analysis predicts transcription factors and paracrine signals that affect fates and experiments validate that the TF Obox6 and the cytokine GDF9 enhance reprogramming efficiency. Our approach sheds light on the process and outcome of reprogramming and provides a framework applicable to diverse temporal processes in biology.
Collapse
Affiliation(s)
- Geoffrey Schiebinger
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; MIT Center for Statistics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jian Shu
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA.
| | - Marcin Tabaka
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Brian Cleary
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Computational and Systems Biology Program, MIT, Cambridge, MA 02142, USA
| | - Vidya Subramanian
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Aryeh Solomon
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Joshua Gould
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Siyan Liu
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Biochemistry Program, Wellesley College, Wellesley, MA 02481, USA
| | - Stacie Lin
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Peter Berube
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Lia Lee
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jenny Chen
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Justin Brumbaugh
- Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Molecular Biology, Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Philippe Rigollet
- MIT Center for Statistics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mathematics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Konrad Hochedlinger
- Department of Molecular Biology, Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Computational and Systems Biology Program, MIT, Cambridge, MA 02142, USA
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| | - Eric S Lander
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Systems Biology Harvard Medical School, Boston, MA 02125, USA.
| |
Collapse
|
26
|
Chen H, Zhang Y, Dai L, Song Y, Wang Y, Zhou B, Zhou R. Association between polymorphisms in CXCR2 gene and preeclampsia. Mol Genet Genomic Med 2019; 7:e00578. [PMID: 30714340 PMCID: PMC6465673 DOI: 10.1002/mgg3.578] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 10/25/2018] [Accepted: 12/28/2018] [Indexed: 12/13/2022] Open
Abstract
Background Preeclampsia is a serious pregnancy‐specific syndrome with incompletely understood pathogenesis. Previous study has demonstrated that the decreased CXCR2 in preeclamptic placentas may contribute to the development of preeclampsia. The role of single nucleotide polymorphisms (SNPs) of CXCR2 gene in the pathogenesis of preeclampsia remains largely unexplored. Thus, we aimed to investigate the association between polymorphisms of CXCR2 gene and preeclampsia in Han Chinese women. Methods Totally 481 pregnant women, including 243 controls and 238 patients with preeclampsia were recruited. The rs1126579 and rs2230054 polymorphisms in CXCR2 gene were tested using polymerase chain reaction‐restriction fragment length polymorphism method. Results Significantly increased risk of preeclampsia was observed in the rs1126579 CC or TC/CC genotypes when compared with TT genotype (CC vs. TT: odss ratio [OR] = 2.11, 95% confidence interval [CI] = 1.18–3.76, p = 0.039; TC/CC vs. TT: OR = 1.89, 95% CI = 1.29–2.78, p = 0.001). Markedly higher risk of preeclampsia was found to be associated with rs1126579 TC genotype (TC vs. TT/CC: OR = 1.48, 95% CI = 1.04–2.12, p = 0.031). After stratification analysis, the different distribution of TC/CC genotypes was particularly significant in the severe preeclampsia group (OR = 2.15, 95% CI = 1.42–3.24, p < 0.01), the early‐onset severe preeclampsia group (OR = 1.97, 95% CI = 1.14–3.42, p = 0.013), and the late‐onset severe preeclampsia group (OR = 2.29, 95% CI = 1.39–3.78, p < 0.01). Besides, TC genotype carriers had a 1.55 fold increased risk of severe preeclampsia (95% CI = 1.06–2.27, p = 0.022) and a 1.80 fold increased risk of late onset severe preeclampsia (95% CI = 1.14–2.83, p = 0.01) than those of TT/CC genotype carriers. Conclusions Our study suggests a genetic association between rs1126579 polymorphism in CXCR2 gene and increased risk of preeclampsia. These data provide a new clue for future investigation.
Collapse
Affiliation(s)
- Hongqin Chen
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, Sichuan, PR China
| | - Yanping Zhang
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, Sichuan, PR China
| | - Li Dai
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, Sichuan, PR China
| | - Yaping Song
- Laboratory of Molecular and Translational Medicine, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, Sichuan, PR China
| | - Yanyun Wang
- Laboratory of Molecular and Translational Medicine, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, Sichuan, PR China
| | - Bin Zhou
- Laboratory of Molecular and Translational Medicine, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, Sichuan, PR China
| | - Rong Zhou
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, Sichuan, PR China
| |
Collapse
|
27
|
Hamilton ST, Hutterer C, Egilmezer E, Steingruber M, Milbradt J, Marschall M, Rawlinson WD. Human cytomegalovirus utilises cellular dual-specificity tyrosine phosphorylation-regulated kinases during placental replication. Placenta 2018; 72-73:10-19. [PMID: 30501876 DOI: 10.1016/j.placenta.2018.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 09/28/2018] [Accepted: 10/12/2018] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Congenital cytomegalovirus (HCMV) infection may cause significant fetal malformation and in severe cases fetal and neonatal death. Fetal injury may be caused indirectly by the placental response to infection. Dual-specificity tyrosine phosphorylation-regulated kinases (DYRKs) have recently been identified as critical kinases for HCMV replication. In this study we provide first evidence that DYRK1A and DYRK1B are utilised during HCMV placental replication. METHODS DYRK expression was investigated in AD169- and Merlin-infected TEV-1 trophoblast cells, ex vivo placental explants and naturally infected clinical placentae by immunofluorescence, western blot, co-immunoprecipitation and RT-qPCR. RESULTS HCMV-infected placental cells showed accumulation and re-localisation of DYRK1A and DYRK1B protein to areas of cytoplasmic virion assembly complexes and nuclear viral replication compartments, respectively. This accumulation was a result of upregulated DYRK1A/B protein expression with HCMV inducing up to a 5.3-fold increase in DYRK1A and up to a 4.7-fold increase in DYRK1B protein, relative to mock-infected TEV-1 cells (p < 0.0001). Increased DYRK protein expression was correlated with DYRK1A/B mRNA upregulation, with HCMV-infected cells showing up to a 3.7-fold increase and 2.9-fold increase in DYRK1A and DYRK1B mRNA levels respectively (p < 0.05). Protein-protein interactions were detected between DYRK1A/1B complexes and HCMV immediate early IE2p86, early pp65 and pUL44 and late pp150 proteins. Treatment of HCMV-infected TEV-1 cells and placental explants with DYRK inhibitors significantly inhibited HCMV replication (p < 0.05) indicating these cellular kinases are required during HCMV placental replication. CONCLUSION HCMV modulates cellular DYRKs during placental replication which may have implications for congenital HCMV pathogenesis and represent promising antiviral targets.
Collapse
Affiliation(s)
- Stuart T Hamilton
- Serology and Virology Division, SEALS Microbiology, Prince of Wales Hospital, Sydney, Australia; School of Women's and Children's Health, University of New South Wales, Australia
| | - Corina Hutterer
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Germany
| | - Ece Egilmezer
- Serology and Virology Division, SEALS Microbiology, Prince of Wales Hospital, Sydney, Australia; School of Medical Sciences, University of New South Wales, Australia
| | - Mirjam Steingruber
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Germany
| | - Jens Milbradt
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Germany
| | - Manfred Marschall
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Germany
| | - William D Rawlinson
- Serology and Virology Division, SEALS Microbiology, Prince of Wales Hospital, Sydney, Australia; School of Women's and Children's Health, University of New South Wales, Australia; School of Medical Sciences, University of New South Wales, Australia; School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia.
| |
Collapse
|
28
|
Zhang SM, Tian FJ, Zeng WH, Ma XL, Ren JB, Lin Y. XCL1-XCR1 pathway promotes trophoblast invasion at maternal-fetal interface by inducing MMP-2/MMP-9 activity. Am J Reprod Immunol 2018; 80:e12990. [PMID: 29856101 DOI: 10.1111/aji.12990] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 05/08/2018] [Indexed: 12/26/2022] Open
Abstract
PROBLEM Certain chemokines with their receptors can promote or inhibit trophoblast cell migration and invasion in human first-trimester placenta. Whether the lymphotactin (Lptn; XCL1)-XC chemokine receptor 1 (XCR1) chemokine pathway affects trophoblast cell migration and invasion in human first-trimester placenta remains unclear. METHOD OF STUDY The expression pattern of chemokine XCL1 and its receptor XCR1 was detected in human first-trimester by qRT-PCR, and the effect of recombinant human XCL1 (rhXCL1) on trophoblast cell function was tested by wound healing and Transwell assays. Matrix metalloproteinase (MMP) activity in trophoblast cells treated with rhXCL1 was assessed via qRT-PCR and gelatin zymography. RESULTS Abundant XCR1 mRNA was expressed in the first-trimester decidua and villi. XCL1 and XCR1 mRNA were expressed at a higher level in the first-trimester than in the term placenta. RhXCL1 promoted trophoblast cell migration and invasion by increasing MMP-9 and MMP-2 activity and that of the MMP-2/tissue inhibitor of metalloproteinases 2 (TIMP-2) complex via the phosphatidylinositol 3-kinase (PI3K)/AKT kinase (AKT), mitogen-activated protein kinase (MEK), and JUN N-terminal kinase (JNK) signaling pathways. CONCLUSION XCL1-XCR1 chemokine pathway promotes trophoblast invasion by increasing matrix metalloproteinase activity in human first-trimester placenta.
Collapse
Affiliation(s)
- Si-Ming Zhang
- Department of Gynecology and Obstetrics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Institute of Embryo-Fetal Original Adult Disease Affiliated to Shanghai Jiao Tong University School of Medicine, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fu-Ju Tian
- Institute of Embryo-Fetal Original Adult Disease Affiliated to Shanghai Jiao Tong University School of Medicine, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-Hong Zeng
- Institute of Embryo-Fetal Original Adult Disease Affiliated to Shanghai Jiao Tong University School of Medicine, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-Ling Ma
- Institute of Embryo-Fetal Original Adult Disease Affiliated to Shanghai Jiao Tong University School of Medicine, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia-Bin Ren
- Department of Gynecology and Obstetrics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Lin
- Institute of Embryo-Fetal Original Adult Disease Affiliated to Shanghai Jiao Tong University School of Medicine, The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
29
|
Wang H, Wang T, Dai L, Cao W, Ye L, Gao L, Zhou B, Zhou R. Effects of CXCL3 on migration, invasion, proliferation and tube formation of trophoblast cells. Placenta 2018; 66:47-56. [PMID: 29884302 DOI: 10.1016/j.placenta.2018.05.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 05/04/2018] [Accepted: 05/10/2018] [Indexed: 01/18/2023]
Abstract
CXCL3 was reportedly associated with the invasion and metastasis of various malignancies, the role of CXCL3, however, in preeclampsia has not been fully discussed. We previously found placental CXCL3 level in severe preeclampsia was significantly lower than that in healthy pregnancy and exogenous recombinant human CXCL3 protein was able to promote trophoblasts' migration and proliferation. The current study, therefore, is further to investigate effects of endogenous CXCL3 on migration, invasion, proliferation, tube formation and apoptosis of trophoblasts. Immunofluorescence staining demonstrated that CXCL3 localized in both trophoblasts of placenta and HTR-8/SVneo cells. Moreover, data showed that migration, invasion, proliferation and tube-formation capability of HTR-8/SVneo cells transfected with siRNA-CXCL3 were suppressed by down-regulation of CXCL3, while those behaviors of HTR-8/SVneo cells transfected with pEZ-CXCL3 were enhanced by upregulation of CXCL3. Nevertheless, the apoptosis of HTR-8/SVneo cells was not affected by neither siRNA nor overexpression plasmid. The result suggests that CXCL3 is involved in migration, invasion, proliferation and tubule formation of trophoblasts and may play a key role in the pathogenesis of preeclampsia.
Collapse
Affiliation(s)
- Hui Wang
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, Sichuan, PR China
| | - Tao Wang
- Laboratory of Molecular and Translational Medicine, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, Sichuan, PR China
| | - Li Dai
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, Sichuan, PR China
| | - Wen Cao
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, Sichuan, PR China
| | - Lei Ye
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, Sichuan, PR China
| | - Linbo Gao
- Laboratory of Molecular and Translational Medicine, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, Sichuan, PR China
| | - Bin Zhou
- Laboratory of Molecular and Translational Medicine, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, Sichuan, PR China
| | - Rong Zhou
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, Chengdu, Sichuan, PR China.
| |
Collapse
|
30
|
Sun M, Na Q, Huang L, Song G, Jin F, Li Y, Hou Y, Kang D, Qiao C. YAP Is Decreased in Preeclampsia and Regulates Invasion and Apoptosis of HTR-8/SVneo. Reprod Sci 2018; 25:1382-1393. [PMID: 29303055 DOI: 10.1177/1933719117746784] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Preeclampsia (PE) is a gestational disorder with hypertension and proteinuria leading to maternal and fetal morbidity and mortality. Yes-associated protein (YAP), a transcription coactivator of Hippo pathway, was identified as an oncoprotein participated in tumorigenesis. However, the effect of YAP on trophoblast has not been investigated. In our study, YAP expression levels in first-trimester, full-term, and PE placentas were detected using quantitative real-time polymerase chain reaction (PCR), Western blot assays, and immunohistochemistry. Yes-associated protein expression was also detected in BeWo and HTR-8/SVneo. Overexpression plasmid and YAP small interfering RNA were introduced into trophoblast cells. Furthermore, we utilized a Transwell invasion assay, flow cytometry, and Cell Counting Kit-8 analysis to examine the role of YAP in the invasion, apoptosis, and proliferation of HTR-8/SVneo trophoblast cells. The result showed that both YAP messenger RNA (mRNA) and protein expression levels were less in preeclamptic placentas. Yes-associated protein mRNA and protein expression levels were more highly expressed in BeWo. Yes-associated protein enhanced cell invasion, reduced the cellular apoptotic response, and had no effect on proliferation. In addition, the overexpression of YAP activated the expression of caudal-related homeobox transcription factor 2 (CDX2), whereas reduced expression of YAP inhibited the expression of CDX2. Our results demonstrate that decreased YAP levels may contribute to the development of PE by regulating trophoblast invasion and apoptosis involving regulation of CDX2. Collectively, we proposed decreased YAP may contribute to trophoblast dysfunction, which suggests it might represent a prognostic biomarker and therapeutic target for PE.
Collapse
Affiliation(s)
- Man Sun
- 1 Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.,2 Key Laboratory of Maternal-Fetal Medicine, China Medical University, Shenyang, Liaoning Province, China.,3 Key Laboratory of Obstetrics and Gynecology of Higher Education, China Medical University, Shenyang, Liaoning Province, China
| | - Quan Na
- 1 Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.,2 Key Laboratory of Maternal-Fetal Medicine, China Medical University, Shenyang, Liaoning Province, China.,3 Key Laboratory of Obstetrics and Gynecology of Higher Education, China Medical University, Shenyang, Liaoning Province, China
| | - Ling Huang
- 1 Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.,2 Key Laboratory of Maternal-Fetal Medicine, China Medical University, Shenyang, Liaoning Province, China.,3 Key Laboratory of Obstetrics and Gynecology of Higher Education, China Medical University, Shenyang, Liaoning Province, China
| | - Guiyu Song
- 1 Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.,2 Key Laboratory of Maternal-Fetal Medicine, China Medical University, Shenyang, Liaoning Province, China.,3 Key Laboratory of Obstetrics and Gynecology of Higher Education, China Medical University, Shenyang, Liaoning Province, China
| | - Feng Jin
- 1 Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.,2 Key Laboratory of Maternal-Fetal Medicine, China Medical University, Shenyang, Liaoning Province, China.,3 Key Laboratory of Obstetrics and Gynecology of Higher Education, China Medical University, Shenyang, Liaoning Province, China
| | - Yuanyuan Li
- 1 Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.,2 Key Laboratory of Maternal-Fetal Medicine, China Medical University, Shenyang, Liaoning Province, China.,3 Key Laboratory of Obstetrics and Gynecology of Higher Education, China Medical University, Shenyang, Liaoning Province, China
| | - Yue Hou
- 1 Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.,2 Key Laboratory of Maternal-Fetal Medicine, China Medical University, Shenyang, Liaoning Province, China.,3 Key Laboratory of Obstetrics and Gynecology of Higher Education, China Medical University, Shenyang, Liaoning Province, China
| | - Danyang Kang
- 1 Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.,2 Key Laboratory of Maternal-Fetal Medicine, China Medical University, Shenyang, Liaoning Province, China.,3 Key Laboratory of Obstetrics and Gynecology of Higher Education, China Medical University, Shenyang, Liaoning Province, China
| | - Chong Qiao
- 1 Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.,2 Key Laboratory of Maternal-Fetal Medicine, China Medical University, Shenyang, Liaoning Province, China.,3 Key Laboratory of Obstetrics and Gynecology of Higher Education, China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
31
|
Li M, Cheng W, Luo J, Hu X, Nie T, Lai H, Zheng X, Li F, Li H. Loss of selenocysteine insertion sequence binding protein 2 suppresses the proliferation, migration/invasion and hormone secretion of human trophoblast cells via the PI3K/Akt and ERK signaling pathway. Placenta 2017. [DOI: 10.1016/j.placenta.2017.05.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
32
|
Fu Y, Wei J, Dai X, Ye Y. Increased NDRG1 expression attenuate trophoblast invasion through ERK/MMP-9 pathway in preeclampsia. Placenta 2017; 51:76-81. [PMID: 28292472 DOI: 10.1016/j.placenta.2017.01.126] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/06/2017] [Accepted: 01/25/2017] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The aim of this study was to investigate the expression of N-myc downstream-regulated gene1(NDRG1)in the placentas of pregnancies complicated with early-onset and late-onset preeclampsia (PE) and its underlying mechanism on the pathophysiology of PE. METHODS The expressions of NDRG-1 in placentas of pregnancies complicated with early-onset PE and late-onset PE were detected using immunohistochemistry, western blot assays and fluorescence quantitative PCR. The expressions of MMP-2, MMP-9 and ERK1/2 protein were detected by western blot analysis and cell invasion assay was performed using transwell chambers in NDRG1 silenced JEG-3 cells. RESULTS Compared with the normal term pregnancies, the expression of both NDRG1 mRNA and protein were significantly high in placentas from PE, and the expression of NDRG1 in early-onset PE was higher than that in late-onset PE. In NDRG1-silenced JEG-3 cells, MMP-2, MMP-9 and phosphorylation of ERK1/2 protein increased obviously and the number of cells that penetrated the membrane increased. CONCLUSION Upregulation of NDRG1 is associated with impaired trophoblast invasion in PE by inhibition ERK/MMP-2 and MMP-9 Pathway.
Collapse
Affiliation(s)
- Yufen Fu
- Department of Obstetrics and Gynecology, Qingdao University, Qingdao 266000, China; Department of Obstetrics, Zibo Maternity and Child Health Hospital, Zibo 255000, China
| | - Jufeng Wei
- Department of Obstetrics and Gynecology, Qingdao University, Qingdao 266000, China; Department of Obstetrics, Qingdao Central Hospital, Qingdao 266000, China
| | - Xueli Dai
- Department of Obstetrics, Zibo Maternity and Child Health Hospital, Zibo 255000, China
| | - Yuanhua Ye
- Department of Obstetrics and Gynecology, Qingdao University, Qingdao 266000, China; Department of Obstetrics, Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| |
Collapse
|