1
|
Chen Y, Xu Y, Zhang Y, Yang D, Sun Y. Functions of the fusogenic and non-fusogenic activities of Syncytin-1 in human physiological and pathological processes. Biochem Biophys Res Commun 2025; 761:151746. [PMID: 40188598 DOI: 10.1016/j.bbrc.2025.151746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/22/2025] [Accepted: 04/01/2025] [Indexed: 04/08/2025]
Abstract
Human endogenous retroviruses (HERVs), which represent the genetic remnants of ancient viral infections, constitute approximately 8 % of the human genome. Among the proteins encoded by these viruses, Syncytin-1, encoded by the env gene of the HERV-W family, functions as a vital fusion protein in placental development, in which it plays a pivotal role in facilitating the fusion of trophoblast cells to form the syncytiotrophoblast that is essential for maintaining the structural integrity and functional viability of the placenta. Recent studies have shown that in addition to its expression in the placenta, Syncytin-1 also plays key roles in a range of different tissues and cell types, influencing biological processes such as cell proliferation, apoptosis, and immune regulation. Abnormal expression of Syncytin-1 has been closely linked to the onset, progression, and metastasis of tumors, potentially promoting tumor invasion via mechanisms involving cell fusion and modulation of the immune microenvironment. Moreover, associations have been established between Syncytin-1 and neurological disorders, including multiple sclerosis and schizophrenia, in which it modulates neuroinflammation. In this review, we systematically examine the molecular structure and functional attributes of Syncytin-1, emphasizing its roles in cell fusion, tumor progression, and immune regulation, and discuss its potential applications as a therapeutic target and diagnostic biomarker.
Collapse
Affiliation(s)
- Yuling Chen
- Medical School, The Affiliated Hospital of Kunming University of Science and Technology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China.
| | - Ya Xu
- Department of Clinical Laboratory, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China.
| | - Yu Zhang
- Department of Clinical Laboratory, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China.
| | - Danni Yang
- Medical School, The Affiliated Hospital of Kunming University of Science and Technology, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China.
| | - Yi Sun
- Institute of Basic and Clinical Medicine, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China.
| |
Collapse
|
2
|
Lestari B, Fukushima T, Utomo RY, Wahyuningsih MSH. Apoptotic and non-apoptotic roles of caspases in placenta physiology and pathology. Placenta 2024; 151:37-47. [PMID: 38703713 DOI: 10.1016/j.placenta.2024.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 03/12/2024] [Accepted: 03/27/2024] [Indexed: 05/06/2024]
Abstract
Caspases, a family of cysteine proteases, are pivotal regulators of apoptosis, the tightly controlled cell death process crucial for eliminating excessive or unnecessary cells during development, including placental development. Collecting research has unveiled the multifaceted roles of caspases in the placenta, extending beyond apoptosis. Apart from their involvement in placental tissue remodeling via apoptosis, caspases actively participate in essential regulatory processes, such as trophoblast fusion and differentiation, significantly influencing placental growth and functionality. In addition, growing evidence indicates an elevation in caspase activity under pathological conditions like pre-eclampsia (PE) and intrauterine growth restriction (IUGR), leading to excessive cell death as well as inflammation. Drawing from advancements in caspase research and placental development under both normal and abnormal conditions, we examine the significance of caspases in both cell death (apoptosis) and non-cell death-related processes within the placenta. We also discuss potential therapeutics targeting caspase-related pathways for placenta disorders.
Collapse
Affiliation(s)
- Beni Lestari
- Department Pharmacology and Therapy, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia; Cancer Chemoprevention Research Center, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Toshiaki Fukushima
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Japan.
| | - Rohmad Yudi Utomo
- Cancer Chemoprevention Research Center, Universitas Gadjah Mada, Yogyakarta, Indonesia; Department Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Mae Sri Hartati Wahyuningsih
- Department Pharmacology and Therapy, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia.
| |
Collapse
|
3
|
Dang Q, Zhu Y, Zhang Y, Hu Z, Wei Y, Chen Z, Jiang X, Cai X, Yu H. Nuclear Binding Protein 2/Nesfatin-1 Affects Trophoblast Cell Fusion during Placental Development via the EGFR-PLCG1-CAMK4 Pathway. Int J Mol Sci 2024; 25:1925. [PMID: 38339201 PMCID: PMC10856506 DOI: 10.3390/ijms25031925] [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: 12/18/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
Previous studies have shown that nuclear binding protein 2 (NUCB2) is expressed in the human placenta and increases with an increase in the syncytialization of trophoblast cells. This study aimed to investigate the role of NUCB2 in the differentiation and fusion of trophectoderm cells. In this study, the expression levels of NUCB2 and E-cadherin in the placentas of rats at different gestation stages were investigated. The results showed that there was an opposite trend between the expression of placental NUCB2 and E-cadherin in rat placentas in different trimesters. When primary human trophoblast (PHT) and BeWo cells were treated with high concentrations of Nesfatin-1, the trophoblast cell syncytialization was significantly inhibited. The effects of NUCB2 knockdown in BeWo cells and Forskolin-induced syncytialization were investigated. These cells showed a significantly decreased cell fusion rate. The mechanism underlying NUCB2-regulated trophoblast cell syncytialization was explored using RNA-Seq and the results indicated that the epidermal growth factor receptor (EGFR)-phospholipase C gamma 1 (PLCG1)-calmodulin-dependent protein kinase IV (CAMK4) pathway might be involved. The results suggested that the placental expression of NUCB2 plays an important role in the fusion of trophoblasts during differentiation via the EGFR-PLCG1-CAMK4 pathway.
Collapse
Affiliation(s)
- Qinyu Dang
- Department of Nutrition and Food Hygiene, School of Public Health, Capital Medical University, Beijing 100069, China; (Q.D.); (Y.Z.); (Y.Z.); (Z.H.); (Y.W.); (Z.C.); (X.C.)
| | - Yandi Zhu
- Department of Nutrition and Food Hygiene, School of Public Health, Capital Medical University, Beijing 100069, China; (Q.D.); (Y.Z.); (Y.Z.); (Z.H.); (Y.W.); (Z.C.); (X.C.)
| | - Yadi Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Capital Medical University, Beijing 100069, China; (Q.D.); (Y.Z.); (Y.Z.); (Z.H.); (Y.W.); (Z.C.); (X.C.)
| | - Zhuo Hu
- Department of Nutrition and Food Hygiene, School of Public Health, Capital Medical University, Beijing 100069, China; (Q.D.); (Y.Z.); (Y.Z.); (Z.H.); (Y.W.); (Z.C.); (X.C.)
| | - Yuchen Wei
- Department of Nutrition and Food Hygiene, School of Public Health, Capital Medical University, Beijing 100069, China; (Q.D.); (Y.Z.); (Y.Z.); (Z.H.); (Y.W.); (Z.C.); (X.C.)
| | - Zhaoyang Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Capital Medical University, Beijing 100069, China; (Q.D.); (Y.Z.); (Y.Z.); (Z.H.); (Y.W.); (Z.C.); (X.C.)
| | - Xinyin Jiang
- Departments of Health and Nutrition Sciences, Brooklyn College of City University of New York, New York, NY 11210, USA;
| | - Xiaxia Cai
- Department of Nutrition and Food Hygiene, School of Public Health, Capital Medical University, Beijing 100069, China; (Q.D.); (Y.Z.); (Y.Z.); (Z.H.); (Y.W.); (Z.C.); (X.C.)
| | - Huanling Yu
- Departments of Health and Nutrition Sciences, Brooklyn College of City University of New York, New York, NY 11210, USA;
| |
Collapse
|
4
|
Abstract
During placentation, villous cytotrophoblast (CTB) stem cells proliferate and fuse, giving rise to the multinucleated syncytiotrophoblast (STB), which represents the terminally differentiated villous layer as well as the maternal-fetal interface. The syncytiotrophoblast is at the forefront of nutrient, gas, and waste exchange while also harboring essential endocrine functions to support pregnancy and fetal development. Considering that mitochondrial dynamics and respiration have been implicated in stem cell fate decisions of several cell types and that the placenta is a mitochondria-rich organ, we will highlight the role of mitochondria in facilitating trophoblast differentiation and maintaining trophoblast function. We discuss both the process of syncytialization and the distinct metabolic characteristics associated with CTB and STB sub-lineages prior to and during syncytialization. As mitochondrial respiration is tightly coupled to redox homeostasis, we emphasize the adaptations of mitochondrial respiration to the hypoxic placental environment. Furthermore, we highlight the critical role of mitochondria in conferring the steroidogenic potential of the STB following differentiation. Ultimately, mitochondrial function and morphological changes centrally regulate respiration and influence trophoblast fate decisions through the production of reactive oxygen species (ROS), whose levels modulate the transcriptional activation or suppression of pluripotency or commitment genes.
Collapse
Affiliation(s)
- Tina Podinić
- Department of Pediatrics and Graduate Program in Medical Sciences, McMaster University, Hamilton, ON, Canada
| | - Andie MacAndrew
- Department of Pediatrics and Graduate Program in Medical Sciences, McMaster University, Hamilton, ON, Canada
| | - Sandeep Raha
- Department of Pediatrics and Graduate Program in Medical Sciences, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
5
|
Komsa-Penkova R, Krumova S, Langari A, Giosheva I, Gartcheva L, Danailova A, Topalova L, Stoyanova T, Strijkova V, Savov A, Todinova S. Blood Plasma Calorimetric Profiles of Women with Preeclampsia: Effect of Oxidative Stress. Antioxidants (Basel) 2023; 12:antiox12051032. [PMID: 37237898 DOI: 10.3390/antiox12051032] [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: 03/31/2023] [Revised: 04/23/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
Preeclampsia is a pregnancy-related disease with poor placentation and presents itself through hypertension and proteinuria. The disease is also associated with the oxidative modification of proteins in maternal blood plasma. In this work, we combine differential scanning calorimetry (DSC), capillary electrophoresis, and atomic force microscopy (AFM) to evaluate the changes in the plasma denaturation profiles of patients with preeclampsia (PE) as compared with those of pregnant controls. Our results demonstrate that the last trimester of pregnancy substantially affects the main calorimetric characteristics of blood plasma from pregnant controls relative to nonpregnant women. These variations correlate well with the changes in protein levels determined by electrophoresis. DSC analysis revealed significant deviations in the plasma heat capacity profiles of preeclamptic patients from those of pregnant controls. These alterations are expressed mainly in a substantial reduction in albumin-assigned transitions and an upward shift in its denaturation temperature, lower calorimetric enthalpy changes, and a reduced ratio of heat capacity in the albumin/globulin-assigned thermal transitions, which are more pronounced in severe PE cases. The in vitro oxidation model shows that the alteration of PE thermograms is partly related to protein oxidation. AFM data detected numerous aggregate formations in the plasma of PE samples and fewer small ones in the pregnant controls, which are not found in healthy nonpregnant samples. These findings could serve as a basis for further investigations to reveal the possible relationship between albumin thermal stabilization, the increased inflammatory state and oxidative stress, and protein misfolding in preeclampsia.
Collapse
Affiliation(s)
- Regina Komsa-Penkova
- Department of Biochemistry, Medical University-Pleven, Sv. Kliment Ohridski Str. 1, 5800 Pleven, Bulgaria
| | - Sashka Krumova
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, "Acad. G. Bontchev" Str. 21, 1113 Sofia, Bulgaria
| | - Ariana Langari
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, "Acad. G. Bontchev" Str. 21, 1113 Sofia, Bulgaria
| | - Ina Giosheva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, "Acad. G. Bontchev" Str. 21, 1113 Sofia, Bulgaria
- National Genetics Laboratory, University Hospital of Obstetrics and Gynecology "Maichin Dom", Medical University Sofia, Zdrave Str. 2, 1431 Sofia, Bulgaria
| | - Lidia Gartcheva
- National Specialized Hospital for Active Treating of Haematological Diseases, 1756 Sofia, Bulgaria
| | - Avgustina Danailova
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, "Acad. G. Bontchev" Str. 21, 1113 Sofia, Bulgaria
| | - Lora Topalova
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, "Acad. G. Bontchev" Str. 21, 1113 Sofia, Bulgaria
| | - Tanya Stoyanova
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, "Acad. G. Bontchev" Str. 21, 1113 Sofia, Bulgaria
| | - Velichka Strijkova
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, "Acad. G. Bontchev" Str. 21, 1113 Sofia, Bulgaria
- Institute of Optical Materials and Technologies "Acad. Yordan Malinovski", Bulgarian Academy of Sciences, "Acad. G. Bontchev" Str. 109, 1113 Sofia, Bulgaria
| | - Alexey Savov
- National Genetics Laboratory, University Hospital of Obstetrics and Gynecology "Maichin Dom", Medical University Sofia, Zdrave Str. 2, 1431 Sofia, Bulgaria
| | - Svetla Todinova
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, "Acad. G. Bontchev" Str. 21, 1113 Sofia, Bulgaria
| |
Collapse
|
6
|
Zhou H, Zhao C, Wang P, Yang W, Zhu H, Zhang S. Regulators involved in trophoblast syncytialization in the placenta of intrauterine growth restriction. Front Endocrinol (Lausanne) 2023; 14:1107182. [PMID: 36798658 PMCID: PMC9927020 DOI: 10.3389/fendo.2023.1107182] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 01/20/2023] [Indexed: 02/04/2023] Open
Abstract
Placental dysfunction refers to the insufficiency of placental perfusion and chronic hypoxia during early pregnancy, which impairs placental function and causes inadequate supply of oxygen and nutrients to the fetus, affecting fetal development and health. Fetal intrauterine growth restriction, one of the most common outcomes of pregnancy-induced hypertensions, can be caused by placental dysfunction, resulting from deficient trophoblast syncytialization, inadequate trophoblast invasion and impaired vascular remodeling. During placental development, cytotrophoblasts fuse to form a multinucleated syncytia barrier, which supplies oxygen and nutrients to meet the metabolic demands for fetal growth. A reduction in the cell fusion index and the number of nuclei in the syncytiotrophoblast are found in the placentas of pregnancies complicated by IUGR, suggesting that the occurrence of IUGR may be related to inadequate trophoblast syncytialization. During the multiple processes of trophoblasts syncytialization, specific proteins and several signaling pathways are involved in coordinating these events and regulating placental function. In addition, epigenetic modifications, cell metabolism, senescence, and autophagy are also involved. Study findings have indicated several abnormally expressed syncytialization-related proteins and signaling pathways in the placentas of pregnancies complicated by IUGR, suggesting that these elements may play a crucial role in the occurrence of IUGR. In this review, we discuss the regulators of trophoblast syncytialization and their abnormal expression in the placentas of pregnancies complicated by IUGR.
Collapse
Affiliation(s)
- Hanjing Zhou
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Chenqiong Zhao
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Peixin Wang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Weijie Yang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Haiyan Zhu
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
- *Correspondence: Songying Zhang, ; Haiyan Zhu,
| | - Songying Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
- *Correspondence: Songying Zhang, ; Haiyan Zhu,
| |
Collapse
|
7
|
Alfian I, Chakraborty A, Yong HEJ, Saini S, Lau RWK, Kalionis B, Dimitriadis E, Alfaidy N, Ricardo SD, Samuel CS, Murthi P. The Placental NLRP3 Inflammasome and Its Downstream Targets, Caspase-1 and Interleukin-6, Are Increased in Human Fetal Growth Restriction: Implications for Aberrant Inflammation-Induced Trophoblast Dysfunction. Cells 2022; 11:1413. [PMID: 35563719 PMCID: PMC9102093 DOI: 10.3390/cells11091413] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/30/2022] [Accepted: 04/08/2022] [Indexed: 02/04/2023] Open
Abstract
Fetal growth restriction (FGR) is commonly associated with placental insufficiency and inflammation. Nonetheless, the role played by inflammasomes in the pathogenesis of FGR is poorly understood. We hypothesised that placental inflammasomes are differentially expressed and contribute to the aberrant trophoblast function. Inflammasome gene expression profiles were characterised by real-time PCR on human placental tissues collected from third trimester FGR and gestation-matched control pregnancies (n = 25/group). The functional significance of a candidate inflammasome was then investigated using lipopolysaccharide (LPS)-induced models of inflammation in human trophoblast organoids, BeWo cells in vitro, and a murine model of FGR in vivo. Placental mRNA expression of NLRP3, caspases 1, 3, and 8, and interleukin 6 increased (>2-fold), while that of the anti-inflammatory cytokine, IL-10, decreased (<2-fold) in FGR compared with control pregnancies. LPS treatment increased NLRP3 and caspase-1 expression (>2-fold) in trophoblast organoids and BeWo cell cultures in vitro, and in the spongiotrophoblast and labyrinth in the murine model of FGR. However, the LPS-induced rise in NLRP3 was attenuated by its siRNA-induced down-regulation in BeWo cell cultures, which correlated with reduced activity of the apoptotic markers, caspase-3 and 8, compared to the control siRNA-treated cells. Our findings support the role of the NLRP3 inflammasome in the inflammation-induced aberrant trophoblast function, which may contribute to FGR.
Collapse
Affiliation(s)
- Irvan Alfian
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia; (I.A.); (A.C.); (S.S.); (R.W.K.L.); (S.D.R.)
- Faculty of Medicine, Universitas Indonesia, Jl. Salemba Raya 6, Jakarta Pusat 10160, Indonesia
| | - Amlan Chakraborty
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia; (I.A.); (A.C.); (S.S.); (R.W.K.L.); (S.D.R.)
| | - Hannah E. J. Yong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore 117609, Singapore;
| | - Sheetal Saini
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia; (I.A.); (A.C.); (S.S.); (R.W.K.L.); (S.D.R.)
| | - Ricky W. K. Lau
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia; (I.A.); (A.C.); (S.S.); (R.W.K.L.); (S.D.R.)
| | - Bill Kalionis
- Department of Maternal-Fetal Medicine Pregnancy Research Centre, The Royal Women’s Hospital, Melbourne, VIC 3052, Australia;
- Department of Obstetrics and Gynaecology, The University of Melbourne, Melbourne, VIC 3052, Australia;
| | - Evdokia Dimitriadis
- Department of Obstetrics and Gynaecology, The University of Melbourne, Melbourne, VIC 3052, Australia;
- Gynaecology Research Centre, The Royal Women’s Hospital, Melbourne, VIC 3052, Australia
| | - Nadia Alfaidy
- Institut National de la Santé et de la Recherche Médicale U1292, Biologie et Biotechnologie pour la Santé, 38043 Grenoble, France;
- Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Biosciences and Biotechnology Institute of Grenoble, 38054 Grenoble, France
- Service Obstétrique & Gynécologie, Centre Hospitalo-Universitaire Grenoble Alpes, University Grenoble-Alpes, CEDEX 9, 38043 Grenoble, France
| | - Sharon D. Ricardo
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia; (I.A.); (A.C.); (S.S.); (R.W.K.L.); (S.D.R.)
| | - Chrishan S. Samuel
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia; (I.A.); (A.C.); (S.S.); (R.W.K.L.); (S.D.R.)
| | - Padma Murthi
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia; (I.A.); (A.C.); (S.S.); (R.W.K.L.); (S.D.R.)
- Department of Maternal-Fetal Medicine Pregnancy Research Centre, The Royal Women’s Hospital, Melbourne, VIC 3052, Australia;
- Department of Obstetrics and Gynaecology, The University of Melbourne, Melbourne, VIC 3052, Australia;
| |
Collapse
|
8
|
Eliesen GA, Fransen M, Kooijman N, van den Broek PH, Russel FG, Greupink R. Effects of tumor necrosis factor on undifferentiated and syncytialised placental choriocarcinoma BeWo cells. Toxicol In Vitro 2022; 80:105327. [DOI: 10.1016/j.tiv.2022.105327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/01/2022] [Indexed: 10/19/2022]
|
9
|
Wang W, Liu J, Pan E. CircHIPK3 contributes to human villous trophoblast growth, migration and invasion via modulating the pathway of miR-346/KCMF1. Placenta 2021; 118:46-54. [PMID: 35032791 DOI: 10.1016/j.placenta.2021.12.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 12/14/2021] [Accepted: 12/23/2021] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Preeclampsia (PE) is one of the leading causes of maternal and perinatal morbidity and mortality worldwide. The regular growth, migration and invasion of villous trophoblast cells contribute to placental development. The objective of this study was to investigate the role and mechanism of circular RNA homeodomain interacting protein kinase 3 (circHIPK3) in the biological functions of trophoblast cells. METHODS The expression of circHIPK3, microRNA-346 (miR-346) and potassium channel modulatory factor 1 (KCMF1) mRNA was measured by quantitative real-time PCR (qPCR). Trophoblast cell proliferation, migration/invasion and cell cycle progression/apoptosis were determined by CCK-8 assay, transwell assay and flow cytometry assay, respectively. The predicted relationship between miR-346 and circHIPK3 or KCMF1 by bioinformatics was confirmed dual-luciferase reporter assay and RIP assay. RESULTS CircHIPK3 and KCMF1 were downregulated, while miR-346 was upregulated in placenta tissues from PE patients. The forced expression of circHIPK3 promoted trophoblast cell proliferation and migration/invasion but alleviated cell cycle arrest and cell apoptosis. MiR-346 was a target of circHIPK3, and miR-346 restoration reversed the effects of circHIPK3 upregulation. In addition, circHIPK3 acted as miR-346 sponge to modulate KCMF1 expression. KCMF1 downregulation partially repressed trophoblast cell proliferation, migration and invasion that were facilitated by miR-346 inhibition or circHIPK3 upregulation. DISCUSSION CircHIPK3 contributes to trophoblast cell proliferation, migration and invasion by upregulating KCMF1 via acting as miR-346 sponge.
Collapse
Affiliation(s)
- Wenxing Wang
- Department of Obstetrical, Yantaishan Hospital, China
| | - Jingying Liu
- Department of Obstetrical, Yantaishan Hospital, China.
| | - Enjie Pan
- Department of Gynecology, Yantai Hospital of Traditional Chinese Medicine, China
| |
Collapse
|
10
|
Could the Human Endogenous Retrovirus-Derived Syncytialization Inhibitor, Suppressyn, Limit Heterotypic Cell Fusion Events in the Decidua? Int J Mol Sci 2021; 22:ijms221910259. [PMID: 34638599 PMCID: PMC8508417 DOI: 10.3390/ijms221910259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 12/29/2022] Open
Abstract
Proper placental development relies on tightly regulated trophoblast differentiation and interaction with maternal cells. Human endogenous retroviruses (HERVs) play an integral role in modulating cell fusion events in the trophoblast cells of the developing placenta. Syncytin-1 (ERVW-1) and its receptor, solute-linked carrier family A member 5 (SLC1A5/ASCT2), promote fusion of cytotrophoblast (CTB) cells to generate the multi-nucleated syncytiotrophoblast (STB) layer which is in direct contact with maternal blood. Another HERV-derived protein known as Suppressyn (ERVH48-1/SUPYN) is implicated in anti-fusogenic events as it shares the common receptor with ERVW-1. Here, we explore primary tissue and publicly available datasets to determine the distribution of ERVW-1, ERVH48-1 and SLC1A5 expression at the maternal-fetal interface. While SLC1A5 is broadly expressed in placental and decidual cell types, ERVW-1 and ERVH48-1 are confined to trophoblast cell types. ERVH48-1 displays higher expression levels in CTB and extravillous trophoblast, than in STB, while ERVW-1 is generally highest in STB. We have demonstrated through gene targeting studies that suppressyn has the ability to prevent ERVW-1-induced fusion events in co-culture models of trophoblast cell/maternal endometrial cell interactions. These findings suggest that differential HERV expression is vital to control fusion and anti-fusogenic events in the placenta and consequently, any imbalance or dysregulation in HERV expression may contribute to adverse pregnancy outcomes.
Collapse
|
11
|
Fan X, Lou J, Zheng X, Wang Y, Wang J, Luo M, Hu M. Interference with lncRNA NEAT1 promotes the proliferation, migration, and invasion of trophoblasts by upregulating miR-411-5p and inhibiting PTEN expression. Immunopharmacol Immunotoxicol 2021; 43:334-342. [PMID: 33876722 DOI: 10.1080/08923973.2021.1910834] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Background: Preeclampsia (PE) is an idiopathic hypertensive disorder of pregnancy, which is related to abnormal placental villi development. Our previous study has found that lncRNA NEAT1 promotes apoptosis of trophoblasts, but the role of NEAT1 in proliferation, migration, and invasion is still unclear. This study explores the role of NEAT1 in proliferation, migration, and invasion of trophoblasts.Methods: NEAT1 and miR-411-5p levels were detected by quantitative real-time PCR. Colony formation assay detected cell proliferation and transwell assay detected cell migration and invasion. Dual-luciferase reporter assay detected the binding between NEAT1 and miR-411-5p as well as the binding between miR-411-5p and PTEN. RNA pull-down assay detected the combination between NEAT1 and miR-411-5p.Result: NEAT1 was increased and miR-411-5p was reduced in PE patients and human trophoblasts (HTR8/SVneo cells) that were induced with H2O2. Interference with NEAT1 promoted cell proliferation, migration, and invasion, and the miR-411-5p inhibitor reversed the effect of siRNA-NEAT1. The expression of PTEN was promoted in PE patients and HTR8/SVneo cells that were induced with H2O2, while the miR-411-5p mimic inhibited PTEN expression, and the plasmid-mediated PTEN overexpression reversed the effect of the miR-411-5p mimic. Besides, under H2O2 induction, the miR-411-5p mimic promoted cell proliferation, migration, and invasion, and the plasmid-mediated PTEN overexpression reversed the effect of the miR-411-5p mimic.Conclusion: Interference with lncRNA NEAT1 promoted the proliferation, migration, and invasion of trophoblasts and alleviated the development of PE, which was partly mediated by upregulating miR-411-5p and inhibiting PTEN expression.
Collapse
Affiliation(s)
- Xufei Fan
- Department of Obstetrics and Gynecology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Jianyi Lou
- Department of Obstetrics and Gynecology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Xiujuan Zheng
- Department of Obstetrics and Gynecology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Ying Wang
- Department of Obstetrics and Gynecology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Jiayue Wang
- Department of Obstetrics and Gynecology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Mengmeng Luo
- Department of Obstetrics and Gynecology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Min Hu
- Department of Obstetrics and Gynecology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| |
Collapse
|
12
|
Yokota R, Bhunu B, Toba H, Intapad S. Sphingolipids and Kidney Disease: Possible Role of Preeclampsia and Intrauterine Growth Restriction (IUGR). KIDNEY360 2021; 2:534-541. [PMID: 35369015 PMCID: PMC8786006 DOI: 10.34067/kid.0006322020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 01/06/2021] [Indexed: 02/04/2023]
Abstract
Sphingolipids are now considered not only as constitutional components of the cellular membrane but also as essential bioactive factors regulating development and physiologic functions. Ceramide is a vital intermediate of sphingolipid metabolism, synthesized by de novo and salvage pathways, producing multiple types of sphingolipids and their metabolites. Although mutations in gene-encoding enzymes regulating sphingolipid synthesis and metabolism cause distinct diseases, an abnormal sphingolipid metabolism contributes to various pathologic conditions, including kidney diseases. Excessive accumulation of glycosphingolipids and promotion of the ceramide salvage and sphingosine-1-phosphate (S1P) pathways are found in the damaged kidney. Acceleration of the sphingosine kinase/S1P/S1P receptor (SphK/S1P/S1PR) axis plays a central role in deteriorating kidney functions. The SphK/S1P/S1PR signaling impairment is also found during pregnancy complications, such as preeclampsia and intrauterine growth restriction (IUGR). This mini-review discusses the current state of knowledge regarding the role of sphingolipid metabolism on kidney diseases, and the possible involvement of preeclampsia and IUGR conditions.
Collapse
Affiliation(s)
- Rodrigo Yokota
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Benjamin Bhunu
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| | - Hiroe Toba
- Division of Pathological Sciences, Department of Clinical Pharmacology, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Suttira Intapad
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| |
Collapse
|
13
|
Colson A, Depoix CL, Baldin P, Hubinont C, Sonveaux P, Debiève F. Hypoxia-inducible factor 2 alpha impairs human cytotrophoblast syncytialization: New insights into placental dysfunction and fetal growth restriction. FASEB J 2020; 34:15222-15235. [PMID: 32954526 DOI: 10.1096/fj.202001681r] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/26/2020] [Accepted: 09/03/2020] [Indexed: 01/01/2023]
Abstract
Insufficient remodeling of uterine arteries causes pregnancy-related diseases, including fetal growth restriction and preeclampsia. In these situations, reduced maternal blood flow in the placenta is thought to be responsible for the persistence of a low oxygen environment throughout pregnancy. We hypothesized that chronic activation of transcription factors hypoxia-inducible factors (HIFs) actively participates in placental underdevelopment, which impairs fetal growth. The computer-assisted analysis in pathological placentas revealed an increased number of HIF-2α-positive nuclei in the syncytium compared to normal human placentas, while HIF-1α stabilization was unchanged. Specific involvement of HIF-2α was confirmed in primary human cytotrophoblasts rendered deficient for HIF1A or HIF2A. Silencing HIF2A increased the expression of main syncytialization markers as well as differentiation and syncytium formation. It also improved placental growth factor bioavailability. None of these changes was seen when silencing HIF1A. Conversely, the experimental induction of HIF-2α expression repressed forskolin-induced differentiation in BeWo choriocarcinoma cells. Our mechanistic insights evidence that transcription factor HIF-2α impairs placental function, thus suggesting its participation in fetal growth restriction and preeclampsia when placentas become chronically hypoxic. Furthermore, it suggests the possibility to develop novel molecular targeting therapies for placental dysfunction.
Collapse
Affiliation(s)
- Arthur Colson
- Pole of Obstetrics, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium.,Pole of Pharmacology & Therapeutics, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| | - Christophe Louis Depoix
- Pole of Obstetrics, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| | - Pamela Baldin
- Department of Pathology, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Corinne Hubinont
- Pole of Obstetrics, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium.,Department of Obstetrics, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology & Therapeutics, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| | - Frédéric Debiève
- Pole of Obstetrics, Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium.,Department of Obstetrics, Cliniques universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
14
|
Siemiątkowska A, Kosicka K, Szpera-Goździewicz A, Krzyścin M, Bręborowicz GH, Główka FK. Cortisol metabolism in pregnancies with small for gestational age neonates. Sci Rep 2019; 9:17890. [PMID: 31784640 PMCID: PMC6884581 DOI: 10.1038/s41598-019-54362-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 11/12/2019] [Indexed: 11/27/2022] Open
Abstract
Small for gestational age (SGA) newborns are often born from hypertensive pregnancies. This study aimed to compare the systemic metabolism of cortisol (F) in pregnancies with SGA and appropriate for gestational age (AGA) infants, considering both the normotensive (NT) and hypertensive patients. We hypothesized that the disturbances in systemic metabolism of F in pre-eclampsia (PE) might be attributed not to hypertension only, but to SGA. The study included 117 pregnants in the third trimester, divided into groups: NT pregnancy and SGA neonate (SGA-NT); NT pregnancy and AGA neonate (AGA-NT; controls), and respective groups with PE: SGA-PE and AGA-PE. We assessed the glucocorticoid balance with the function of enzymes involved in systemic metabolism of F: 11β-hydroxysteroid dehydrogenase type 1 and 2 (11β-HSD1 and 11β-HSD2), 5α- and 5β-reductase. The enzymes' functions were estimated with the levels of F, cortisone (E), and their metabolites in plasma or urine, which we measured with HPLC-FLD and HPLC-MS/MS. The plasma F/E and urinary free F/E (UFF/UFE) ratios correlated significantly only in patients with the normal function of 5α- and 5β-reductase. The increased function of 11β-HSD2 was noted in all pre-eclamptic pregnancies. Increased function of 5α- and 5β-reductase was specific only for SGA-PE pregnancies, and the function of 5α-reductase was dependent on fetal sex. The SGA-NT pregnancies with male fetuses trended towards the higher function of renal 11β-HSD2 and 5β-reductase; SGA-NT pregnancies with female fetuses lacked any systemic glucocorticoid imbalance. In conclusion, systemic metabolism of F is the most intensive in pre-eclamptic pregnancies complicated by SGA with female fetuses. Our study supports the hypothesis about the different origins of PE and idiopathic intrauterine growth restriction and suggests the sex-specific mechanisms responsible for fetal growth restriction.
Collapse
Affiliation(s)
- Anna Siemiątkowska
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, 6 Święcickiego Street, 60-781, Poznań, Poland
| | - Katarzyna Kosicka
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, 6 Święcickiego Street, 60-781, Poznań, Poland.
| | - Agata Szpera-Goździewicz
- Department of Perinatology and Gynecology, Poznan University of Medical Sciences, 33 Polna Street, 60-535, Poznań, Poland
| | - Mariola Krzyścin
- Department of Perinatology and Gynecology, Poznan University of Medical Sciences, 33 Polna Street, 60-535, Poznań, Poland
| | - Grzegorz H Bręborowicz
- Department of Perinatology and Gynecology, Poznan University of Medical Sciences, 33 Polna Street, 60-535, Poznań, Poland
| | - Franciszek K Główka
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, 6 Święcickiego Street, 60-781, Poznań, Poland
| |
Collapse
|
15
|
Lappas M, McCracken S, McKelvey K, Lim R, James J, Roberts CT, Fournier T, Alfaidy N, Powell KL, Borg AJ, Morris JM, Leaw B, Singh H, Ebeling PR, Wallace EM, Parry LJ, Dimitriadis E, Murthi P. Formyl peptide receptor-2 is decreased in foetal growth restriction and contributes to placental dysfunction. Mol Hum Reprod 2019; 24:94-109. [PMID: 29272530 DOI: 10.1093/molehr/gax067] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 12/18/2017] [Indexed: 01/07/2023] Open
Abstract
STUDY QUESTION What is the association between placental formyl peptide receptor 2 (FPR2) and trophoblast and endothelial functions in pregnancies affected by foetal growth restriction (FGR)? SUMMARY ANSWER Reduced FPR2 placental expression in idiopathic FGR results in significantly altered trophoblast differentiation and endothelial function in vitro. WHAT IS KNOWN ALREADY FGR is associated with placental insufficiency, where defective trophoblast and endothelial functions contribute to reduced feto-placental growth. STUDY DESIGN, SIZE, DURATION The expression of FPR2 in placental tissues from human pregnancies complicated with FGR was compared to that in gestation-matched uncomplicated control pregnancies (n = 25 from each group). Fpr2 expression was also determined in placental tissues obtained from a murine model of FGR (n = 4). The functional role of FPR2 in primary trophoblasts and endothelial cells in vitro was assessed in diverse assays in a time-dependent manner. PARTICIPANTS/MATERIALS, SETTING, METHODS Placentae from third-trimester pregnancies complicated by idiopathic FGR (n = 25) and those from gestation-matched pregnancies with appropriately grown infants as controls (n = 25) were collected at gestation 27-40 weeks. Placental tissues were also collected from a spontaneous CBA/CaH × DBA/2 J murine model of FGR. Placental FPR2/Fpr2 mRNA expression was determined by real-time PCR, while protein expression was examined by immunoblotting and immunohistochemistry. siRNA transfection was used to silence FPR2 expression in primary trophoblasts and in human umbilical vein endothelial cells (HUVEC), and the quantitation of cytokines, chemokines and apoptosis was performed following a cDNA array analyses. Functional effects of trophoblast differentiation were measured using HCGB/β-hCG and syncytin-2 expression as well as markers of apoptosis, tumour protein 53 (TP53), caspase 8, B cell lymphoma 2 (BCL2) and BCL associated X (BAX). Endothelial function was assessed by proliferation, network formation and permeability assays. MAIN RESULTS AND THE ROLE OF CHANCE Placental FPR2/Fpr2 expression was significantly decreased in FGR placentae (n = 25, P < 0.05) as well as in murine FGR placentae compared to controls (n = 4, P < 0.05). FPR2 siRNA (siFPR2) in term trophoblasts significantly increased differentiation markers, HCGB and syncytin-2; cytokines, interleukin (IL)-6, CXCL8; and apoptotic markers, TP53, caspase 8 and BAX, but significantly reduced the expression of the chemokines CXCL12 and its receptors CXCR4 and CXCR7; CXCL16 and its receptor, CXCR6; and cytokine, IL-10, compared with control siRNA (siCONT). Treatment of HUVECs with siFPR2 significantly reduced proliferation and endothelial tube formation, but significantly increased permeability of HUVECs. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Reduced expression of placental FPR2/Fpr2 was observed in the third trimester at delivery after development of FGR, suggesting that FPR2 is associated with FGR pregnancies. However, there is a possibility that the decreased placental FPR2 observed in FGR may be a consequence rather than a cause of FGR, although our in vitro functional analyses using primary trophoblasts and endothelial cells suggest that FPR2 may have a direct or indirect regulatory role on trophoblast differentiation and endothelial function in FGR. WIDER IMPLICATIONS OF THE FINDINGS This is the first study linking placental FPR2 expression with changes in the trophoblast and endothelial functions that may explain the placental insufficiency observed in FGR. STUDY FUNDING/COMPETING INTERESTS P.M. and P.R.E. received funding from the Australian Institute of Musculoskeletal Science, Western Health, St. Albans, Victoria 3021, Australia. M.L. is supported by a Career Development Fellowship from the National Health and Medical Research Council (NHMRC; Grant no. 1047025). Monash Health is supported by the Victorian Government's Operational Infrastructure Support Programme. The authors declare that there is no conflict of interest in publishing this work.
Collapse
Affiliation(s)
- Martha Lappas
- Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Victoria 3079, Australia.,Department of Obstetrics and Gynaecology, University of Melbourne, Heidelberg, Victoria 3079, Australia
| | - Sharon McCracken
- Division of Perinatal Research, Kolling Institute, Northern Sydney Local Health District, St Leonards, New South Wales 2065, Australia.,Sydney Medical School Northern, University of Sydney, New South Wales 2006, Australia
| | - Kelly McKelvey
- Division of Perinatal Research, Kolling Institute, Northern Sydney Local Health District, St Leonards, New South Wales 2065, Australia.,Sydney Medical School Northern, University of Sydney, New South Wales 2006, Australia
| | - Ratana Lim
- Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Victoria 3079, Australia.,Department of Obstetrics and Gynaecology, University of Melbourne, Heidelberg, Victoria 3079, Australia
| | - Joanna James
- Department of Obstetrics and Gynaecology, University of Auckland, New Zealand
| | - Claire T Roberts
- Adelaide Medical School and Robinson Research Institute, University of Adelaide, South Australia 5005, Australia
| | - Thierry Fournier
- INSERM, UMR-S1139, Faculté des Sciences Pharmaceutiques et Biologiques, Paris F-75006, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris F-75006 France.,Fondation PremUp, Paris F-75006, France
| | - Nadia Alfaidy
- Institut National de la Santé et de la Recherche Médicale, Unité 1036, Grenoble, France.,University Grenoble-Alpes, 38000 Grenoble, France.,Commissariat à l'Energie Atomique (CEA), iRTSV- Biology of Cancer and Infection, Grenoble, France
| | - Katie L Powell
- Division of Perinatal Research, Kolling Institute, Northern Sydney Local Health District, St Leonards, New South Wales 2065, Australia.,Sydney Medical School Northern, University of Sydney, New South Wales 2006, Australia
| | - Anthony J Borg
- Department of Maternal-Fetal Medicine Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria 3052, Australia
| | - Jonathan M Morris
- Division of Perinatal Research, Kolling Institute, Northern Sydney Local Health District, St Leonards, New South Wales 2065, Australia.,Sydney Medical School Northern, University of Sydney, New South Wales 2006, Australia
| | - Bryan Leaw
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Harmeet Singh
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Peter R Ebeling
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia
| | - Euan M Wallace
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.,Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia
| | - Laura J Parry
- School of Biosciences, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Evdokia Dimitriadis
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Padma Murthi
- Department of Maternal-Fetal Medicine Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria 3052, Australia.,The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.,Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia.,Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria 3052, Australia
| |
Collapse
|
16
|
Human Placenta Expresses α 2-Adrenergic Receptors and May Be Implicated in Pathogenesis of Preeclampsia and Fetal Growth Restriction. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2774-2785. [PMID: 30273604 DOI: 10.1016/j.ajpath.2018.08.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/07/2018] [Accepted: 08/23/2018] [Indexed: 02/02/2023]
Abstract
α2-Adrenergic receptors (α2ARs) are G-protein-coupled receptors involved in catecholamine signaling by extracellular regulated protein kinase 1 and 2 (ERK1/2) pathways. We examined placental expression and function of α2AR subtypes in women with severe preeclampsia (sPE) with and without intrauterine growth restriction (IUGR). Placental biopsies were analyzed from 52 women with i) sPE (n = 8); ii) sPE + IUGR (n = 9); iii) idiopathic IUGR (n = 8); iv) idiopathic preterm birth (n = 16); and v) healthy term controls (n = 11). Expression of α2AR subtypes (α2A, α2B, α2C) and phospho-ERK1/2 (receptor activation marker) was investigated by immunohistochemistry and/or quantitative real-time RT-PCR. The effects of α2CAR knockdown on syncytialization (syncytin-1 and -2) and β-human chorionic gonadotropin secretion were examined in BeWo cells stimulated with forskolin. The effects of α2AR agonist UK 14,304 and specific α2CAR antagonist were tested, using a trophoblast migration assay. All three α2ARs were expressed and functionally active in human placenta with site-specific localization. Highest α2BAR and α2CAR mRNA expression was identified in sPE + IUGR. α2CAR knockdown increased expression of syncytin-1 and -2 but decreased secretion of β-human chorionic gonadotropin. UK 14,304 impaired trophoblast migration. The observed α2AR expression pattern suggests different function for each subtype. α2CAR modulates trophoblast syncytialization and migration and may carry pathogenic role in sPE + IUGR.
Collapse
|
17
|
Ravikumar G, Crasta J, Prabhu JS, Thomas T, Dwarkanath P, Thomas A, Kurpad AV, Sridhar TS. CD15 as a marker of fetoplacental endothelial immaturity in IUGR placentas. J Matern Fetal Neonatal Med 2017; 32:1646-1653. [DOI: 10.1080/14767058.2017.1414179] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Gayatri Ravikumar
- Department of Pathology, St. John’s Medical College, Koramangala, Bangalore, India
| | - Julian Crasta
- Department of Pathology, St. John’s Medical College, Koramangala, Bangalore, India
| | - Jyothi S. Prabhu
- Division of Molecular Medicine, St. John’s Research Institute, Koramangala, Bangalore, India
| | - Tinku Thomas
- Department of Biostatistics, St. John’s Medical College, Koramangala, Bangalore, India
| | - Pratibha Dwarkanath
- Division of Nutrition, St. John’s Research Institute, Koramangala, Bangalore, India
| | - Annamma Thomas
- Department of Obstetrics and Gynaecology, St. John’s Medical College Hospital, Koramangala, Bangalore, India
| | - Anura V. Kurpad
- Department of Physiology, Division of Nutrition, St. John’s Medical College, Koramangala, Bangalore, India
| | | |
Collapse
|
18
|
Huebner H, Hartner A, Rascher W, Strick RR, Kehl S, Heindl F, Wachter DL, Beckmann Md MW, Fahlbusch FB, Ruebner M. Expression and Regulation of Retinoic Acid Receptor Responders in the Human Placenta. Reprod Sci 2017; 25:1357-1370. [PMID: 29246089 DOI: 10.1177/1933719117746761] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Retinoic acid (RA) signaling through its receptors (RARA, RARB, RARG, and the retinoic X receptor RXRA) is essential for healthy placental and fetal development. An important group of genes regulated by RA are the RA receptor responders (RARRES1, 2, and 3). We set out to analyze their expression and regulation in healthy and pathologically altered placentas of preeclampsia (PE) and intrauterine growth restriction (IUGR) as well as in trophoblast cell lines. METHODS We performed immunohistochemical staining on placental sections and analyzed gene expression by real-time polymerase chain reaction. Additionally, we performed cell culture experiments and stimulated Swan71 and Jeg-3 cells with different RA derivates and 2'-deoxy-5-azacytidine (AZA) to induce DNA demethylation. RESULTS RARRES1, 2, and 3 and RARA, RARB, RARG, and RXRA are expressed in the extravillous part of the placenta. RARRES1, RARA, RARG, and RXRA were additionally detected in villous cytotrophoblasts. RARRES gene expression was induced via activation of RARA, RARB, and RARG in trophoblast cells. RARRES1 was overexpressed in villous trophoblasts and the syncytiotrophoblast from PE placentas, but not in IUGR without PE. Promoter methylation was detectable for RARRES1 and RARB based on their sensitivity toward AZA treatment of trophoblast cell lines. DISCUSSION RARRES1, 2 and 3 are expressed in the functional compartments of the human placenta and can be regulated by RA. We hypothesize that the epigenetic suppression of trophoblast RARRES1 and RARB expression and the upregulation of RARRES1 in PE trophoblast cells suggest an involvement of environmental factors (eg, maternal vitamin A intake) in the pathogenesis of this pregnancy complication.
Collapse
Affiliation(s)
- Hanna Huebner
- 1 Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Andrea Hartner
- 2 Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Wolfgang Rascher
- 2 Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Reiner R Strick
- 1 Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Sven Kehl
- 1 Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Felix Heindl
- 1 Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - David L Wachter
- 3 Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany * The authors are contributed equally
| | - Matthias W Beckmann Md
- 1 Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Fabian B Fahlbusch
- 2 Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander-University of Erlangen-Nuremberg, Erlangen, Germany
| | - Matthias Ruebner
- 1 Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
19
|
Knabl J, Vattai A, Ye Y, Jueckstock J, Hutter S, Kainer F, Mahner S, Jeschke U. Role of Placental VDR Expression and Function in Common Late Pregnancy Disorders. Int J Mol Sci 2017; 18:ijms18112340. [PMID: 29113124 PMCID: PMC5713309 DOI: 10.3390/ijms18112340] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 10/24/2017] [Accepted: 10/26/2017] [Indexed: 12/14/2022] Open
Abstract
Vitamin D, besides its classical role in bone metabolism, plays a distinct role in multiple pathways of the feto-maternal unit. Calcitriol is the major active ligand of the nuclear vitamin D receptor (VDR). The vitamin D receptor (VDR) is expressed in different uteroplacental parts and exerts a variety of functions in physiologic pregnancy. It regulates decidualisation and implantation, influences hormone secretion and placental immune modulations. This review highlights the role of the vitamin D receptor in physiologic and disturbed pregnancy, as preeclampsia, fetal growth restriction, gestational diabetes and preterm birth. We discuss the existing literature regarding common VDR polymorphisms in these pregnancy disorders.
Collapse
Affiliation(s)
- Julia Knabl
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians Universität München, 80337 Munich, Germany.
- Department of Obstetrics and Gynecology, Klinik Hallerwiese, 90419 Nürnberg, Germany.
| | - Aurelia Vattai
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians Universität München, 80337 Munich, Germany.
| | - Yao Ye
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians Universität München, 80337 Munich, Germany.
| | - Julia Jueckstock
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians Universität München, 80337 Munich, Germany.
| | - Stefan Hutter
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians Universität München, 80337 Munich, Germany.
| | - Franz Kainer
- Department of Obstetrics and Gynecology, Klinik Hallerwiese, 90419 Nürnberg, Germany.
| | - Sven Mahner
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians Universität München, 80337 Munich, Germany.
| | - Udo Jeschke
- Department of Obstetrics and Gynecology, University Hospital, Ludwig-Maximilians Universität München, 80337 Munich, Germany.
| |
Collapse
|
20
|
Zadora J, Singh M, Herse F, Przybyl L, Haase N, Golic M, Yung HW, Huppertz B, Cartwright JE, Whitley G, Johnsen GM, Levi G, Isbruch A, Schulz H, Luft FC, Müller DN, Staff AC, Hurst LD, Dechend R, Izsvák Z. Disturbed Placental Imprinting in Preeclampsia Leads to Altered Expression of DLX5, a Human-Specific Early Trophoblast Marker. Circulation 2017; 136:1824-1839. [PMID: 28904069 PMCID: PMC5671803 DOI: 10.1161/circulationaha.117.028110] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 08/28/2017] [Indexed: 01/23/2023]
Abstract
Supplemental Digital Content is available in the text. Background: Preeclampsia is a complex and common human-specific pregnancy syndrome associated with placental pathology. The human specificity provides both intellectual and methodological challenges, lacking a robust model system. Given the role of imprinted genes in human placentation and the vulnerability of imprinted genes to loss of imprinting changes, there has been extensive speculation, but no robust evidence, that imprinted genes are involved in preeclampsia. Our study aims to investigate whether disturbed imprinting contributes to preeclampsia. Methods: We first aimed to confirm that preeclampsia is a disease of the placenta by generating and analyzing genome-wide molecular data on well-characterized patient material. We performed high-throughput transcriptome analyses of multiple placenta samples from healthy controls and patients with preeclampsia. Next, we identified differentially expressed genes in preeclamptic placentas and intersected them with the list of human imprinted genes. We used bioinformatics/statistical analyses to confirm association between imprinting and preeclampsia and to predict biological processes affected in preeclampsia. Validation included epigenetic and cellular assays. In terms of human specificity, we established an in vitro invasion-differentiation trophoblast model. Our comparative phylogenetic analysis involved single-cell transcriptome data of human, macaque, and mouse preimplantation embryogenesis. Results: We found disturbed placental imprinting in preeclampsia and revealed potential candidates, including GATA3 and DLX5, with poorly explored imprinted status and no prior association with preeclampsia. As a result of loss of imprinting, DLX5 was upregulated in 69% of preeclamptic placentas. Levels of DLX5 correlated with classic preeclampsia markers. DLX5 is expressed in human but not in murine trophoblast. The DLX5high phenotype resulted in reduced proliferation, increased metabolism, and endoplasmic reticulum stress-response activation in trophoblasts in vitro. The transcriptional profile of such cells mimics the transcriptome of preeclamptic placentas. Pan-mammalian comparative analysis identified DLX5 as part of the human-specific regulatory network of trophoblast differentiation. Conclusions: Our analysis provides evidence of a true association among disturbed imprinting, gene expression, and preeclampsia. As a result of disturbed imprinting, the upregulated DLX5 affects trophoblast proliferation. Our in vitro model might fill a vital niche in preeclampsia research. Human-specific regulatory circuitry of DLX5 might help explain certain aspects of preeclampsia.
Collapse
Affiliation(s)
- Julianna Zadora
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Manvendra Singh
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Florian Herse
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Lukasz Przybyl
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Nadine Haase
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Michaela Golic
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Hong Wa Yung
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Berthold Huppertz
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Judith E Cartwright
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Guy Whitley
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Guro M Johnsen
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Giovanni Levi
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Annette Isbruch
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Herbert Schulz
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Friedrich C Luft
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Dominik N Müller
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Anne Cathrine Staff
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.)
| | - Laurence D Hurst
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.).
| | - Ralf Dechend
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.).
| | - Zsuzsanna Izsvák
- From Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (J.Z., M.S., F.H., N.H., D.N.M., Z.I.); Experimental and Clinical Research Center, a joint cooperation between the Max-Delbrück Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Germany (J.Z., F.H., L.P., N.H., M.G., H.S., F.C.L., D.N.M., R.D.); Berlin Institute of Health, Germany (J.Z., F.H., L.P., N.H., M.G., F.C.L., D.N.M., R.D., Z.I.); Department of Obstetrics and Department of Gynecology, Charité-Universitätsmedizin Berlin, Germany (M.G.); German Centre for Cardiovascular Research, partner site Berlin, Germany (N.H., D.N.M.); Centre for Trophoblast Research, University of Cambridge, UK (H.W.Y.); Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Austria (B.H.); Molecular and Clinical Sciences Research Institute, St George's University of London, UK (J.E.C., G.W.); Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (G.M.J., A.C.S.); University of Oslo, Norway (G.M.J., A.C.S.); Évolution des Régulations Endocriniennes, Muséum Nationale d'Histoire Naturelle, Paris, France (G.L.); HELIOS-Klinikum, Berlin, Germany (A.I., R.D.); Cologne Center for Genomics, University of Cologne, Germany (H.S.); and Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, UK (L.D.H.).
| |
Collapse
|
21
|
Plasma cross-gestational sphingolipidomic analyses reveal potential first trimester biomarkers of preeclampsia. PLoS One 2017; 12:e0175118. [PMID: 28384202 PMCID: PMC5383057 DOI: 10.1371/journal.pone.0175118] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 03/21/2017] [Indexed: 01/01/2023] Open
Abstract
Introduction Preeclampsia (PE) is a gestational disorder, manifested in the second half of pregnancy by maternal hypertension, proteinuria and generalized edema. PE is a major cause of maternal and fetal morbidity and mortality, accounting for nearly 40% of all premature births worldwide. Bioactive sphingolipids are emerging as key molecules involved in etiopathogenesis of PE, characterized by maternal angiogenic imbalance and symptoms of metabolic syndrome. The aim of this study was to compare the cross-gestational profile of circulating bioactive sphingolipids in maternal plasma from preeclamptic (PE) versus normotensive control (CTL) subjects with the goal of identifying sphingolipids as candidate first trimester biomarkers of PE for early prediction of the disease. Methods A prospective cohort of patients was sampled at the first, second and third trimester of pregnancy for each patient (11–14, 22–24, and 32–36 weeks´ gestation). A retrospective stratified study design was used to quantify different classes of sphingolipids in maternal plasma. We used a reverse-phase high-performance liquid chromatography-tandem mass spectrometry (HPLC-ESI-MS/MS) approach for determining different sphingolipid molecular species (sphingosine-1-phosphate (S1P), dihydro-sphingosine-1-phosphate (DH-S1P), sphingomyelins (SM) and ceramides (Cer)) in cross-gestational samples of human plasma from PE (n = 7, 21 plasma samples across pregnancy) and CTL (n = 7, 21 plasma samples across pregnancy) patients. Results Plasma levels of angiogenic S1P did not change significantly in control and in preeclamptic patients´ group across gestation. DH-S1P was significantly decreased in second trimester plasma of PE patients in comparison to their first trimester, which could contribute to reduced endothelial barrier observed in PE. The major ceramide species (Cer 16:0 and Cer 24:0) tended to be up-regulated in plasma of control and PE subjects across gestation. The levels of a less abundant plasma ceramide species (Cer 14:0) were significantly lower in first trimester plasma of PE patients when compared with their gestational-matched control samples (p = 0.009). Major plasma sphingomyelin species (SM 16:0, SM 18:1 and SM 24:0) tended to be higher in control pregnancies across gestation. However, in PE patients, SM 16:0, SM 18:0 and SM 18:1 showed significant up-regulation across gestation, pointing to atherogenic properties of the sphingomyelins and particularly the potential contribution of SM 18:0 to the disease development. In addition, two major sphingomyelins, SM 16:0 and SM 18:0, were significantly lower in first trimester plasma of PE patients versus first trimester samples of respective controls (p = 0.007 and p = 0.002, respectively). Conclusions Cross-gestational analysis of maternal plasma of preeclamptic and normotensive women identifies differences in the biochemical profile of major sphingolipids (DH-S1P, sphingomyelins and ceramides) between these two groups. In addition, first trimester maternal plasma sphingolipids (Cer 14:0, SM 16:0 and SM 18:0) may serve in the future as early biomarkers of PE occurrence and development.
Collapse
|
22
|
p45 NF-E2 regulates syncytiotrophoblast differentiation by post-translational GCM1 modifications in human intrauterine growth restriction. Cell Death Dis 2017; 8:e2730. [PMID: 28383551 PMCID: PMC5477575 DOI: 10.1038/cddis.2017.127] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 02/03/2017] [Accepted: 02/08/2017] [Indexed: 01/21/2023]
Abstract
Placental insufficiency jeopardizes prenatal development, potentially leading to intrauterine growth restriction (IUGR) and stillbirth. Surviving fetuses are at an increased risk for chronic diseases later in life. IUGR is closely linked with altered trophoblast and placental differentiation. However, due to a paucity of mechanistic insights, suitable biomarkers and specific therapies for IUGR are lacking. The transcription factor p45 NF-E2 (nuclear factor erythroid derived 2) has been recently found to regulate trophoblast differentiation in mice. The absence of p45 NF-E2 in trophoblast cells causes IUGR and placental insufficiency in mice, but mechanistic insights are incomplete and the relevance of p45 NF-E2 for human syncytiotrophoblast differentiation remains unknown. Here we show that p45 NF-E2 negatively regulates human syncytiotrophoblast differentiation and is associated with IUGR in humans. Expression of p45 NF-E2 is reduced in human placentae complicated with IUGR compared with healthy controls. Reduced p45 NF-E2 expression is associated with increased syncytiotrophoblast differentiation, enhanced glial cells missing-1 (GCM1) acetylation and GCM1 desumoylation in IUGR placentae. Induction of syncytiotrophoblast differentiation in BeWo and primary villous trophoblast cells with 8-bromo-adenosine 3',5'-cyclic monophosphate (8-Br-cAMP) reduces p45 NF-E2 expression. Of note, p45 NF-E2 knockdown is sufficient to increase syncytiotrophoblast differentiation and GCM1 expression. Loss of p45 NF-E2 using either approach resulted in CBP-mediated GCM1 acetylation and SENP-mediated GCM1 desumoylation, demonstrating that p45 NF-E2 regulates post-translational modifications of GCM1. Functionally, reduced p45 NF-E2 expression is associated with increased cell death and caspase-3 activation in vitro and in placental tissues samples. Overexpression of p45 NF-E2 is sufficient to repress GCM1 expression, acetylation and desumoylation, even in 8-Br-cAMP exposed BeWo cells. These results suggest that p45 NF-E2 negatively regulates differentiation and apoptosis activation of human syncytiotrophoblast by modulating GCM1 acetylation and sumoylation. These studies identify a new pathomechanism related to IUGR in humans and thus provide new impetus for future studies aiming to identify new biomarkers and/or therapies of IUGR.
Collapse
|
23
|
Alan B, Tunç S, Agacayak E, Bilici A. Diagnosis of pre-eclampsia and assessment of severity through examination of the placenta with acoustic radiation force impulse elastography. Int J Gynaecol Obstet 2016; 135:43-6. [PMID: 27452611 DOI: 10.1016/j.ijgo.2016.03.037] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 03/22/2016] [Accepted: 06/24/2016] [Indexed: 12/09/2022]
Abstract
OBJECTIVE To assess the value of placental shear wave velocity (SWV) measurement by acoustic radiation force impulse (ARFI) imaging for the diagnosis of pre-eclampsia and to determine the relationship between the SWV and the severity of pre-eclampsia. METHODS A prospective study was performed at a center in Turkey between August 2014 and March 2015. The study included consecutive pregnant women in the second or third trimester diagnosed with pre-eclampsia and healthy pregnant women without pre-eclampsia of similar ages. Patients with pre-eclampsia were divided into two groups (severe or mild disease) on the basis of revised American College of Obstetricians and Gynecologists criteria. All patients underwent ARFI, and the SWV was measured at several placental locations. RESULTS Overall, 86 women were enrolled (42 with pre-eclampsia, 44 controls). Minimum, maximum, and mean SWV values were significantly higher in the pre-eclampsia group than in the control group (P<0.001 for all). These values were also significantly higher among patients with severe pre-eclampsia than among patients with mild pre-eclampsia (P<0.001 for all). CONCLUSION Measurement of the placental SWV with ARFI imaging is a useful additional method for the diagnosis of pre-eclampsia and for determination of the disease severity.
Collapse
Affiliation(s)
- Bircan Alan
- Department of Radiology, Dicle University Medical Faculty, Diyarbakır, Turkey.
| | - Senem Tunç
- Department of Gynecology and Obstetrics, Dicle University Medical Faculty, Diyarbakır, Turkey
| | - Elif Agacayak
- Department of Gynecology and Obstetrics, Dicle University Medical Faculty, Diyarbakır, Turkey
| | - Aslan Bilici
- Department of Radiology, Dicle University Medical Faculty, Diyarbakır, Turkey
| |
Collapse
|
24
|
Weber M, Göhner C, San Martin S, Vattai A, Hutter S, Parraga M, Jeschke U, Schleussner E, Markert UR, Fitzgerald JS. Unique trophoblast stem cell- and pluripotency marker staining patterns depending on gestational age and placenta-associated pregnancy complications. Cell Adh Migr 2016; 10:56-65. [PMID: 26914354 DOI: 10.1080/19336918.2016.1142035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Preeclampsia (PE) and intrauterine growth retardation (IUGR) are rare but severe pregnancy complications that are associated with placental insufficiency often resulting in premature birth. The clinical pathologies are related to gross placental pathologies and trophoblastic deficiencies that might derive from inflammatory processes and oxidative stress injury. The mesenchymal core of placental villi has been identified as a possible niche for trophoblast progenitor cells that are called upon to replenish the injured syncytiotrophoblast layer. These progenitor cells are known to express trophoblast stem cell (CDX2) and pluripotency (SOX2, NANOG and OCT4A) markers, however only little data is available characterizing the expression of these transcription factors beyond the blastocyst stage. We aimed to describe the expression of these factors in healthy 1st and 3rd trimester placentae as well as PE, IUGR and combined PE+IUGR placentae. We analyzed 8 respective samples derived from 1st trimester (elective abortions), and 3rd trimester (healthy controls, PE, IUGR and combined PE+IUGR). We accomplished immunoperoxidase staining to detect the stem cell markers: CDX2 (trophectoderm), SOX2, NANOG and OCT4A (embryonal). Immunoreative scoring was used for objective analyses of staining patterns. All markers display clearly elevated signals in 1st trimester villous samples as compared to healthy 3rd trimester counterparts. Especially CDX2 and NANOG were specific to the cytotrophoblast layer and the mesenchymal core. Specific and differential expression patterns were visible in the villous/extravillous compartment of each placenta-associated pregnancy complication (PE: pan elevated expression; IUGR elevated SOX2 in basal plate; combined PE+IUGR pan loss of expression). Reduction of stem cell transcription factor expression in term placentae indicates temporal regulation, and probably a specific function which is yet to be elucidated. The differential expression patterns within placentae complicated with placenta-associated pregnancy complications indicate that PE, IUGR and combined PE+IUGR are separate entities. It is unclear whether the alterations are the cause or the effect of the clinical pathology.
Collapse
Affiliation(s)
- Maja Weber
- a Department of Obstetrics , Placenta Lab, University Hospital Jena , Jena , Germany
| | - Claudia Göhner
- a Department of Obstetrics , Placenta Lab, University Hospital Jena , Jena , Germany.,b Department of Obstetrics and Gynecology , University Medical Center Groningen, University of Groningen , Groningen , The Netherlands
| | - Sebastian San Martin
- c Biomedical Research Center, School of Medicine, Universidad de Valparaiso , Chile
| | - Aurelia Vattai
- d Department of Obstetrics and Gynecology , Ludwig Maximilians University of Munich , Munich , Germany
| | - Stefan Hutter
- d Department of Obstetrics and Gynecology , Ludwig Maximilians University of Munich , Munich , Germany
| | - Mario Parraga
- c Biomedical Research Center, School of Medicine, Universidad de Valparaiso , Chile
| | - Udo Jeschke
- d Department of Obstetrics and Gynecology , Ludwig Maximilians University of Munich , Munich , Germany
| | - Ekkehard Schleussner
- a Department of Obstetrics , Placenta Lab, University Hospital Jena , Jena , Germany
| | - Udo R Markert
- a Department of Obstetrics , Placenta Lab, University Hospital Jena , Jena , Germany
| | - Justine S Fitzgerald
- a Department of Obstetrics , Placenta Lab, University Hospital Jena , Jena , Germany.,e Praxisklinik am Anger, Kinderwunschzentrum Erfurt , Erfurt , Germany
| |
Collapse
|
25
|
Murthi P, Yong HEJ, Ngyuen TPH, Ellery S, Singh H, Rahman R, Dickinson H, Walker DW, Davies-Tuck M, Wallace EM, Ebeling PR. Role of the Placental Vitamin D Receptor in Modulating Feto-Placental Growth in Fetal Growth Restriction and Preeclampsia-Affected Pregnancies. Front Physiol 2016; 7:43. [PMID: 26924988 PMCID: PMC4757640 DOI: 10.3389/fphys.2016.00043] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 02/01/2016] [Indexed: 12/31/2022] Open
Abstract
Fetal growth restriction (FGR) is a common pregnancy complication that affects up to 5% of pregnancies worldwide. Recent studies demonstrate that Vitamin D deficiency is implicated in reduced fetal growth, which may be rescued by supplementation of Vitamin D. Despite this, the pathway(s) by which Vitamin D modulate fetal growth remains to be investigated. Our own studies demonstrate that the Vitamin D receptor (VDR) is significantly decreased in placentae from human pregnancies complicated by FGR and contributes to abnormal placental trophoblast apoptosis and differentiation and regulation of cell-cycle genes in vitro. Thus, Vitamin D signaling is important for normal placental function and fetal growth. This review discusses the association of Vitamin D with fetal growth, the function of Vitamin D and its receptor in pregnancy, as well as the functional significance of a placental source of Vitamin D in FGR. Additionally, we propose that for Vitamin D to be clinically effective to prevent and manage FGR, the molecular mechanisms of Vitamin D and its receptor in modulating fetal growth requires further investigation.
Collapse
Affiliation(s)
- Padma Murthi
- Department of Medicine, School of Clinical Sciences, Monash UniversityMelbourne, VIC, Australia; Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash UniversityMelbourne, VIC, Australia; Department of Obstetrics and Gynaecology, The University of MelbourneMelbourne, VIC, Australia; Department of Maternal-Fetal Medicine Pregnancy Research Centre, The Royal Women's HospitalMelbourne, VIC, Australia; The Ritchie Centre, Hudson Institute of Medical ResearchMelbourne, VIC, Australia
| | - Hannah E J Yong
- Department of Obstetrics and Gynaecology, The University of MelbourneMelbourne, VIC, Australia; Department of Maternal-Fetal Medicine Pregnancy Research Centre, The Royal Women's HospitalMelbourne, VIC, Australia
| | - Thy P H Ngyuen
- Department of Obstetrics and Gynaecology, The University of MelbourneMelbourne, VIC, Australia; Department of Maternal-Fetal Medicine Pregnancy Research Centre, The Royal Women's HospitalMelbourne, VIC, Australia
| | - Stacey Ellery
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash UniversityMelbourne, VIC, Australia; The Ritchie Centre, Hudson Institute of Medical ResearchMelbourne, VIC, Australia
| | - Harmeet Singh
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash UniversityMelbourne, VIC, Australia; The Ritchie Centre, Hudson Institute of Medical ResearchMelbourne, VIC, Australia
| | - Rahana Rahman
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash UniversityMelbourne, VIC, Australia; The Ritchie Centre, Hudson Institute of Medical ResearchMelbourne, VIC, Australia
| | - Hayley Dickinson
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash UniversityMelbourne, VIC, Australia; The Ritchie Centre, Hudson Institute of Medical ResearchMelbourne, VIC, Australia
| | - David W Walker
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash UniversityMelbourne, VIC, Australia; The Ritchie Centre, Hudson Institute of Medical ResearchMelbourne, VIC, Australia
| | - Miranda Davies-Tuck
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash UniversityMelbourne, VIC, Australia; The Ritchie Centre, Hudson Institute of Medical ResearchMelbourne, VIC, Australia
| | - Euan M Wallace
- Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash UniversityMelbourne, VIC, Australia; The Ritchie Centre, Hudson Institute of Medical ResearchMelbourne, VIC, Australia
| | - Peter R Ebeling
- Department of Medicine, School of Clinical Sciences, Monash University Melbourne, VIC, Australia
| |
Collapse
|
26
|
Roland CS, Hu J, Ren CE, Chen H, Li J, Varvoutis MS, Leaphart LW, Byck DB, Zhu X, Jiang SW. Morphological changes of placental syncytium and their implications for the pathogenesis of preeclampsia. Cell Mol Life Sci 2016; 73:365-376. [PMID: 26496726 PMCID: PMC4846582 DOI: 10.1007/s00018-015-2069-x] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Revised: 09/29/2015] [Accepted: 10/12/2015] [Indexed: 02/05/2023]
Abstract
Preeclampsia is a hypertensive disease that complicates many pregnancies, typically presenting with new-onset or worsening hypertension and proteinuria. It is well recognized that the placental syncytium plays a key role in the pathogenesis of preeclampsia. This review summarizes the findings pertaining to the structural alterations in the syncytium of preeclamptic placentas and analyzes their pathological implications for the development of preeclampsia. Changes in the trophoblastic lineage, including those in the proliferation of cytotrophoblasts, the formation of syncytiotrophoblast through cell fusion, cell apoptosis and syncytial deportation, are discussed in the context of preeclampsia. Extensive correlations are made between functional deficiencies and the alterations on the levels of gross anatomy, tissue histology, cellular events, ultrastructure, molecular pathways, and gene expression. Attention is given to the significance of dynamic changes in the syncytial turnover in preeclamptic placentas. Specifically, experimental evidences for the complex and obligatory role of syncytin-1 in cell fusion, cell-cycle regulation at the G1/S transition, and apoptosis through AIF-mediated pathway, are discussed in detail in the context of syncytium homeostasis. Finally, the recent observations on the aberrant fibrin deposition in the trophoblastic layer and the trophoblast immature phenotype in preeclamptic placentas and their potential pathogenic impact are also reviewed.
Collapse
Affiliation(s)
- Cynthia S Roland
- Department of Obstetrics and Gynecology, Memorial Health University Medical Center, Savannah, GA, 31404, USA
| | - Jian Hu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Chun-E Ren
- Department of Obstetrics and Gynecology, Center of Reproductive Medicine, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Haibin Chen
- Department of Histology and Embryology, Shantou University Medical College, Shantou, Guangdong, China
| | - Jinping Li
- Department of Obstetrics and Gynecology, Memorial Health University Medical Center, Savannah, GA, 31404, USA
- Department of Biomedical Science, Mercer University School of Medicine, Savannah, GA, 31404, USA
| | - Megan S Varvoutis
- Department of Obstetrics and Gynecology, Memorial Health University Medical Center, Savannah, GA, 31404, USA
| | - Lynn W Leaphart
- Department of Obstetrics and Gynecology, Memorial Health University Medical Center, Savannah, GA, 31404, USA
| | - David B Byck
- Department of Obstetrics and Gynecology, Memorial Health University Medical Center, Savannah, GA, 31404, USA
| | - Xueqiong Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
| | - Shi-Wen Jiang
- Department of Obstetrics and Gynecology, Memorial Health University Medical Center, Savannah, GA, 31404, USA.
- Department of Biomedical Science, Mercer University School of Medicine, Savannah, GA, 31404, USA.
| |
Collapse
|
27
|
Higgins LE, Rey de Castro N, Addo N, Wareing M, Greenwood SL, Jones RL, Sibley CP, Johnstone ED, Heazell AEP. Placental Features of Late-Onset Adverse Pregnancy Outcome. PLoS One 2015; 10:e0129117. [PMID: 26120838 PMCID: PMC4488264 DOI: 10.1371/journal.pone.0129117] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 05/05/2015] [Indexed: 12/12/2022] Open
Abstract
Objective Currently, no investigations reliably identify placental dysfunction in late pregnancy. To facilitate the development of such investigations we aimed to identify placental features that differ between normal and adverse outcome in late pregnancy in a group of pregnancies with reduced fetal movement. Methods Following third trimester presentation with reduced fetal movement (N = 100), placental structure ex vivo was measured. Placental function was then assessed in terms of (i) chorionic plate artery agonist responses and length-tension characteristics using wire myography and (ii) production and release of placentally derived hormones (by quantitative polymerase chain reaction and enzyme linked immunosorbant assay of villous tissue and explant conditioned culture medium). Results Placentas from pregnancies ending in adverse outcome (N = 23) were ~25% smaller in weight, volume, length, width and disc area (all p<0.0001) compared with those from normal outcome pregnancies. Villous and trophoblast areas were unchanged, but villous vascularity was reduced (median (interquartile range): adverse outcome 10 (10–12) vessels/mm2 vs. normal outcome 13 (12–15), p = 0.002). Adverse outcome pregnancy placental arteries were relatively insensitive to nitric oxide donated by sodium nitroprusside compared to normal outcome pregnancy placental arteries (50% Effective Concentration 30 (19–50) nM vs. 12 (6–24), p = 0.02). Adverse outcome pregnancy placental tissue contained less human chorionic gonadotrophin (20 (11–50) vs. 55 (24–102) mIU/mg, p = 0.007) and human placental lactogen (11 (6–14) vs. 27 (9–50) mg/mg, p = 0.006) and released more soluble fms-like tyrosine kinase-1 (21 (13–29) vs. 5 (2–15) ng/mg, p = 0.01) compared with normal outcome pregnancy placental tissue. Conclusion These data provide a description of the placental phenotype of adverse outcome in late pregnancy. Antenatal tests that accurately reflect elements of this phenotype may improve its prediction.
Collapse
Affiliation(s)
- Lucy E. Higgins
- Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, Manchester, M13 9WL, United Kingdom
- Maternal and Fetal Health Research Centre, St. Mary's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, United Kingdom
- * E-mail:
| | - Nicolas Rey de Castro
- Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, Manchester, M13 9WL, United Kingdom
- Maternal and Fetal Health Research Centre, St. Mary's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, United Kingdom
| | - Naa Addo
- Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, Manchester, M13 9WL, United Kingdom
- Maternal and Fetal Health Research Centre, St. Mary's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, United Kingdom
| | - Mark Wareing
- Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, Manchester, M13 9WL, United Kingdom
- Maternal and Fetal Health Research Centre, St. Mary's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, United Kingdom
| | - Susan L. Greenwood
- Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, Manchester, M13 9WL, United Kingdom
- Maternal and Fetal Health Research Centre, St. Mary's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, United Kingdom
| | - Rebecca L. Jones
- Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, Manchester, M13 9WL, United Kingdom
- Maternal and Fetal Health Research Centre, St. Mary's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, United Kingdom
| | - Colin P. Sibley
- Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, Manchester, M13 9WL, United Kingdom
- Maternal and Fetal Health Research Centre, St. Mary's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, United Kingdom
| | - Edward D. Johnstone
- Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, Manchester, M13 9WL, United Kingdom
- Maternal and Fetal Health Research Centre, St. Mary's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, United Kingdom
| | - Alexander E. P. Heazell
- Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, Manchester, M13 9WL, United Kingdom
- Maternal and Fetal Health Research Centre, St. Mary's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, United Kingdom
| |
Collapse
|
28
|
Nguyen TPH, Yong HEJ, Chollangi T, Borg AJ, Brennecke SP, Murthi P. Placental vitamin D receptor expression is decreased in human idiopathic fetal growth restriction. J Mol Med (Berl) 2015; 93:795-805. [PMID: 25716068 DOI: 10.1007/s00109-015-1267-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 02/13/2015] [Accepted: 02/16/2015] [Indexed: 01/09/2023]
Abstract
UNLABELLED Fetal growth restriction (FGR) affects up to 5 % of pregnancies worldwide, and trophoblast function plays a significant role on the outcome. An epidemiological study has linked vitamin D deficiency to adverse perinatal outcomes, which include decreased birth weight. The placenta as an important source of vitamin D regulates its metabolism through the vitamin D receptor (VDR), but the mechanism by which VDR regulates trophoblast function is poorly understood. Our study aimed at determining placental VDR expression in FGR and gestation-matched control (GMC) pregnancies and identifying the actions of VDR in trophoblast differentiation and apoptosis. Placentae were collected from a well-defined cohort of idiopathic FGR and GMC pregnancies. VDR mRNA and protein expressions were determined by PCR, immunohistochemistry and immunoblotting, while functional consequences of VDR inactivation in vitro were determined on BeWo cells by determining changes in differentiation, attachment and apoptosis. Significant decreases in VDR mRNA expression (p = 0.0005) and protein expression (p = 0.0003) were observed in the FGR samples, while VDR inactivation, which showed markers for differentiation, cell attachment and apoptosis, was significantly increased. Thus, decreased placental VDR may contribute to uncontrolled premature differentiation and apoptosis of trophoblasts that are characteristics of idiopathic FGR pregnancies. KEY MESSAGE Fetal growth restriction (FGR) affects up to 5 % of all pregnancies worldwide. FGR is the second highest cause of perinatal mortality and morbidity. The placenta plays a pivotal role in vitamin D metabolism during pregnancy. Vitamin D deficiency is associated with adverse pregnancy outcomes. Placental vitamin D receptor expression is decreased in FGR.
Collapse
Affiliation(s)
- T P H Nguyen
- Department of Perinatal Medicine Pregnancy Research Centre, The University of Melbourne, Melbourne, Australia
| | | | | | | | | | | |
Collapse
|
29
|
Riddell MR, Winkler-Lowen B, Jiang Y, Guilbert LJ, Davidge ST. Fibrocyte-like cells from intrauterine growth restriction placentas have a reduced ability to stimulate angiogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:1025-33. [PMID: 23835310 DOI: 10.1016/j.ajpath.2013.06.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Revised: 05/27/2013] [Accepted: 06/04/2013] [Indexed: 10/26/2022]
Abstract
Intrauterine growth restriction (IUGR) is a common complication of pregnancy whereby the fetus fails to achieve its genetic growth potential. Malformation of the placental vasculature is observed in IUGR and may be due to the development of the placenta in a chronically hypoxic environment. Recently, we identified that the predominant stromal cells in the angiogenic zones of the placenta are fibrocyte-like cells. The conditioned medium from fibrocyte-like cells (FcCM) has been shown to stimulate angiogenesis in vitro. Thus, we hypothesized that FcCM from IUGR cells would have a reduced ability to stimulate angiogenesis and that chronic hypoxia would decrease the ability of both normal and IUGR fibrocyte-like cells to stimulate angiogenesis. IUGR FcCM had a reduced ability to stimulate endothelial tubule-like structure formation and an increased ability to stimulate endothelial migration compared with normal FcCM. However, normal and IUGR FcCM produced in chronic hypoxia did not alter endothelial proliferation, migration, or tubule-like structure formation. IUGR FcCM was found to have reduced levels of the pro-angiogenic cytokine IL-8 and increased levels of the anti-angiogenic factors activin-A and pigment epithelium-derived growth factor. Thus, alterations in the ability of IUGR fibrocyte-like cells to stimulate angiogenesis may contribute to the development of vascular malformation in IUGR, but in vitro these changes cannot be attributed to a chronically hypoxic environment.
Collapse
Affiliation(s)
- Meghan R Riddell
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | |
Collapse
|
30
|
Pathirage NA, Cocquebert M, Sadovsky Y, Abumaree M, Manuelpillai U, Borg A, Keogh RJ, Brennecke SP, Evain-Brion D, Fournier T, Kalionis B, Murthi P. Homeobox gene transforming growth factor β-induced factor-1 (TGIF-1) is a regulator of villous trophoblast differentiation and its expression is increased in human idiopathic fetal growth restriction. Mol Hum Reprod 2013; 19:665-75. [PMID: 23761267 DOI: 10.1093/molehr/gat042] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Abnormal trophoblast function is associated with human fetal growth restriction (FGR). Targeted disruption of homeobox gene transforming growth β-induced factor (TGIF-1) results in placental dysfunction in the mouse. The role of human TGIF-1 in placental cell function is unknown. The aims of this study were to determine the expression of TGIF-1 in human idiopathic FGR-affected placentae compared with gestation-matched controls (GMC), to elucidate the functional role of TGIF-1 in trophoblasts and to identify its downstream targets. Real-time PCR and immunoblotting revealed that TGIF-1 mRNA and protein expression was significantly increased in FGR-affected placentae compared with GMC (n = 25 in each group P < 0.05). Immunoreactive TGIF-1 was localized to the villous cytotrophoblasts, syncytiotrophoblast, microvascular endothelial cells and in scattered stromal cells in both FGR and GMC. TGIF-1 inactivation in BeWo cells using two independent siRNA resulted in significantly decreased mRNA and protein of trophoblast differentiation markers, human chorionic gonadotrophin (CGB/hCG), syncytin and 3β-hydroxysteroid dehydrogenase/3β-honest significant difference expression. Our data demonstrate that homeobox gene TGIF-1 is a potential up-stream regulator of trophoblast differentiation and the altered TGIF-1 expression may contribute to aberrant villous trophoblast differentiation in FGR.
Collapse
Affiliation(s)
- Niroshani A Pathirage
- Department of Perinatal Medicine Pregnancy Research Centre, and University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Locked Bag 300, Parkville, VIC 3052, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Hiden U, Ghaffari-Tabrizi N, Gauster M, Tam-Amersdorfer C, Cetin I, Dieber-Rotheneder M, Lang U, Desoye G. Membrane-type matrix metalloproteinase 1 regulates trophoblast functions and is reduced in fetal growth restriction. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:1563-71. [PMID: 23470162 DOI: 10.1016/j.ajpath.2013.01.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 01/07/2013] [Accepted: 01/14/2013] [Indexed: 01/10/2023]
Abstract
Fetal growth restriction (FGR) results from placental insufficiency to adequately supply the fetus. This insufficiency involves impaired cytotrophoblast functions, including reduced migration and invasion, proliferation, and syncytium formation. Membrane-type matrix metalloproteinase 1 (MT1-MMP) is a key enzyme in these cellular processes. MT1-MMP exists in various forms: a 63-kDa proenzyme is synthesized as primary translation product, which is cleaved into a 57-kDa membrane-anchored active form. We hypothesized that reduced placental MT1-MMP in FGR impairs trophoblast functions. MT1-MMP mRNA and active enzyme was quantified in placentas from FGR and age-matched control pregnancies. MT1-MMP protein was localized in first-trimester and term placentas. Putative MT1-MMP functions in trophoblasts were determined using two blocking antibodies for measuring migration and proliferation, as well as fusion of primary trophoblasts and trophoblast-derived cells. MT1-MMP was expressed predominantly in the syncytiotrophoblast and the villous and extravillous cytotrophoblasts. In FGR placentas, levels of MT1-MMP mRNA and of active MT1-MMP protein were reduced (-34.2%, P < 0.05, and -21.5%, P < 0.01, respectively), compared with age-matched controls. MT1-MMP-blocking antibodies diminished migration, proliferation, and trophoblast fusion. We conclude that reduced placental MT1-MMP in FGR may contribute to the impaired trophoblast functions associated with this pathology.
Collapse
Affiliation(s)
- Ursula Hiden
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Singh AT, Dharmarajan A, Aye ILMH, Keelan JA. Ceramide biosynthesis and metabolism in trophoblast syncytialization. Mol Cell Endocrinol 2012; 362:48-59. [PMID: 22652149 DOI: 10.1016/j.mce.2012.05.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2011] [Revised: 05/02/2012] [Accepted: 05/18/2012] [Indexed: 02/02/2023]
Abstract
Sphingolipid mediators such as ceramide are pleiotropic regulators of cellular growth, differentiation and apoptosis. We investigated the role of ceramide biosynthesis, metabolism and actions in term human cytotrophoblasts syncytialized over 7 days in culture. Intracellular C16 ceramide levels increased modestly after 3 days in culture, then declined. Ceramidase was present at particularly high levels in syncytialized trophoblasts; inhibition of ceramidase reduced the degree of cell fusion. Exposure to short chain C8 ceramide or aSMase enhanced secretion of the differentiation marker hCG without affecting fusion or cell viability. In contrast, pharmacological inhibition of ceramidase reduced the extent of fusion. Inhibition of the ceramide-responsive JNK and PP2A pathways did not abolish the effects of ceramide, and JNK phosphorylation was unresponsive to ceramide; however, ceramide significantly inhibited phosphorylation of Akt. This study suggests that changes in ceramide biosynthesis and metabolism play a differential role in the biochemical and morphological features of trophoblast differentiation.
Collapse
Affiliation(s)
- Ambika T Singh
- School of Women's and Infant's Health, Dentistry and Health Sciences, The University of Western Australia, Perth, Australia
| | | | | | | |
Collapse
|
33
|
Männik J, Vaas P, Rull K, Teesalu P, Laan M. Differential placental expression profile of human Growth Hormone/Chorionic Somatomammotropin genes in pregnancies with pre-eclampsia and gestational diabetes mellitus. Mol Cell Endocrinol 2012; 355:180-7. [PMID: 22387044 PMCID: PMC3325480 DOI: 10.1016/j.mce.2012.02.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Revised: 02/09/2012] [Accepted: 02/09/2012] [Indexed: 12/23/2022]
Abstract
The human GH/CSH cluster consisting of one pituitary-expressed (GH1) and four placenta-expressed loci has been implicated in maternal metabolic adaptation to pregnancy, regulation of intrauterine and postnatal growth. We investigated how the mRNA expression profile of placental GH2, CSH1 and CSH2 genes and their alternative transcripts correlates with maternal pre-eclampsia (PE) and/or gestational diabetes mellitus (GD). The expression of studied genes in PE placentas (n=17) compared to controls (n=17) exhibited a trend for reduced transcript levels. The alternative transcripts retaining intron 4, GH2-2 and CSH1-2 showed significantly reduced expression in PE cases without growth restriction (P=0.007, P=0.008, respectively). In maternal GD (n=23), a tendency of differential expression was detected only for the GH2 gene and in pregnancies with large-for-gestational-age newborns. Our results, together with those reported by others, are consistent with a pleiotropic effect of placental hGH/CSH genes at the maternal-fetal interface relating to the regulation of fetal growth and the risk of affected maternal metabolism.
Collapse
Affiliation(s)
- Jaana Männik
- Human Molecular Genetics Group, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia
| | - Pille Vaas
- Human Molecular Genetics Group, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia
- Women’s Clinic of Tartu University Hospital, L. Puusepa 8, 51014 Tartu, Estonia
| | - Kristiina Rull
- Human Molecular Genetics Group, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia
- Women’s Clinic of Tartu University Hospital, L. Puusepa 8, 51014 Tartu, Estonia
| | - Pille Teesalu
- Women’s Clinic of Tartu University Hospital, L. Puusepa 8, 51014 Tartu, Estonia
| | - Maris Laan
- Human Molecular Genetics Group, Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia
| |
Collapse
|
34
|
Riddell MR, Winkler-Lowen B, Chakrabarti S, Dunk C, Davidge ST, Guilbert LJ. The characterization of fibrocyte-like cells: a novel fibroblastic cell of the placenta. Placenta 2012; 33:143-50. [PMID: 22230251 DOI: 10.1016/j.placenta.2011.12.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Revised: 12/13/2011] [Accepted: 12/15/2011] [Indexed: 11/25/2022]
Abstract
The placenta is a highly vascularized organ thus angiogenesis is a key process in placental development. The contribution that different cells in the villous stroma play in placental angiogenesis is largely unknown. In this study we identified a novel stromal cell type in sections of term placenta which is morphologically fibroblastic and expressing the fibroblast marker TE-7 but also positive for the monocytic markers CD115 and CD14 and designated these cells as fibrocyte-like cells. Populations of fibrocyte-like cells from the placenta were isolated by two methods: culture of adherence-selected placental cells and, for higher purity, by CD45 fluorescence activated cell sorting (FACS). Fibrocyte-like cell conditioned medium increased endothelial tubule-like structure formation 2-fold versus control medium. Both pro-angiogenic growth factors vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (b-FGF) and the anti-angiogenic factor soluble-Flt were found in the conditioned medium. Neutralizing antibodies against VEGF and b-FGF reduced endothelial cell tubule-like structures to control levels. These data suggests that fibrocyte-like cells, a previously unidentified cell of the villous stroma, may play an important role in the regulation of placental angiogenesis.
Collapse
Affiliation(s)
- M R Riddell
- Department of Physiology, University of Alberta, Edmonton, Canada T6G 2S2
| | | | | | | | | | | |
Collapse
|
35
|
Homeobox gene Distal-Less 3 is a regulator of villous cytotrophoblast differentiation and its expression is increased in human idiopathic foetal growth restriction. J Mol Med (Berl) 2011; 90:273-84. [DOI: 10.1007/s00109-011-0836-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 10/18/2011] [Accepted: 11/03/2011] [Indexed: 10/15/2022]
|
36
|
Association of placental inflammation with fetomaternal hemorrhage and loss of placental mucin-1. Arch Gynecol Obstet 2011; 285:605-12. [PMID: 21805141 DOI: 10.1007/s00404-011-2028-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2010] [Accepted: 07/16/2011] [Indexed: 01/04/2023]
Abstract
BACKGROUND Fetomaternal hemorrhage (FMH) poses an immediate risk to the fetus and, in case of Rhesus-immunization, to future pregnancies. Given that altered endothelial permeability is part of the pathophysiology of inflammation, in this study we investigated whether placental inflammatory processes like chorioamnionitis (ChoA) or preeclampsia (PE) lead to increased rates of FMH compared to the established risk factor of placenta previa (PP). Putative accompanying markers of trophoblastic damage were also explored. METHODS 40 patients (14 PE; 6 ChoA; 9 PP; 11 normal controls) were evaluated for FMH using a flowcytometric test kit, which is able to quantify FMH of 0.06% fetal cells. Placental tissue samples were immunostained for human placental lactogen (hPL), human chorionic gonadotropin (hCG), and mucin-1 (MUC1). MUC1 was evaluated as a potential serum marker of FMH. RESULTS Patients with ChoA had a mean calculated FMH volume of 29 ml, compared to 4 ml in PE and 1 ml in PP and controls. MUC1 staining was reduced in PE and ChoA placenta samples, while elevated MUC1 serum concentration correlated positively with FMH. CONCLUSION Diseases of placental inflammation are associated with FMH. Placental MUC1 staining is reduced and serum concentrations are increased in cases of FMH.
Collapse
|
37
|
Laskowska M, Laskowska K, Oleszczuk J. Soluble CD30 in normotensive pregnant women with isolated fetal intrauterine growth restriction: a comparison with preeclamptic women. J Reprod Immunol 2010; 86:122-5. [DOI: 10.1016/j.jri.2010.05.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2009] [Revised: 04/19/2010] [Accepted: 05/16/2010] [Indexed: 11/16/2022]
|
38
|
Vasilopoulou E, Loubière LS, Martín-Santos A, McCabe CJ, Franklyn JA, Kilby MD, Chan SY. Differential triiodothyronine responsiveness and transport by human cytotrophoblasts from normal and growth-restricted pregnancies. J Clin Endocrinol Metab 2010; 95:4762-70. [PMID: 20660035 PMCID: PMC3050105 DOI: 10.1210/jc.2010-0354] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
CONTEXT Abnormal placentation in human pregnancy is associated with intrauterine fetal growth restriction (IUGR). Our group has previously reported the association between severe IUGR, lower fetal circulating concentrations of thyroid hormones (THs), and altered expression of TH receptors and TH transporters within human placental villi. We postulate that altered TH bioavailability to trophoblasts may contribute to the pathogenesis of IUGR. DESIGN AND OBJECTIVE Cytotrophoblasts were isolated from normal and IUGR human placentae to compare their responsiveness to T(3) and their capability for T(3) transport. RESULTS Compared with normal cytotrophoblasts, the viability of IUGR cytotrophoblasts (assessed by methyltetrazoleum assay) was significantly reduced (P < 0.001), whereas apoptosis (assessed using caspase 3/7 activity and M30 immunoreactivity) was significantly increased after T(3) treatment for 48 h (P < 0.001 and P < 0.01, respectively). The secretion of human chorionic gonadotropin was significantly increased by IUGR cytotrophoblasts compared with normal cytotrophoblasts (P < 0.001), independently of T(3) treatment. Net transport of [(125)I]T(3) was 20% higher by IUGR cytotrophoblasts compared with normal cytotrophoblasts (P < 0.001), and this was accompanied by a 2-fold increase in the protein expression of the TH transporter, monocarboxylate transporter 8, as assessed by Western immunoblotting (P < 0.01). CONCLUSIONS IUGR cytotrophoblasts demonstrate altered responsiveness to T(3) with significant effects on cell survival and apoptosis compared with normal cytotrophoblasts. Increased monocarboxylate transporter 8 expression and intracellular T(3) accumulation may contribute to the altered T(3) responsiveness of IUGR cytotrophoblasts.
Collapse
Affiliation(s)
- E Vasilopoulou
- School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
39
|
James JL, Whitley GS, Cartwright JE. Pre-eclampsia: fitting together the placental, immune and cardiovascular pieces. J Pathol 2010; 221:363-78. [PMID: 20593492 DOI: 10.1002/path.2719] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The success of pregnancy is a result of countless ongoing interactions between the placenta and the maternal immune and cardiovascular systems. Pre-eclampsia is a serious pregnancy complication that arises from multiple potential aberrations in these systems. The pathophysiology of pre-eclampsia is established in the first trimester of pregnancy, when a range of deficiencies in placentation affect the key process of spiral artery remodelling. As pregnancy progresses to the third trimester, inadequate spiral artery remodelling along with multiple haemodynamic, placental and maternal factors converge to activate the maternal immune and cardiovascular systems, events which may in part result from increased shedding of placental debris. As we understand more about the pathophysiology of pre-eclampsia, it is becoming clear that the development of early- and late-onset pre-eclampsia, as well as intrauterine growth restriction (IUGR), does not necessarily arise from the same underlying pathology.
Collapse
Affiliation(s)
- Joanna L James
- Division of Basic Medical Sciences, St George's University of London, London, UK.
| | | | | |
Collapse
|
40
|
Impaired cell fusion and differentiation in placentae from patients with intrauterine growth restriction correlate with reduced levels of HERV envelope genes. J Mol Med (Berl) 2010; 88:1143-56. [PMID: 20664994 DOI: 10.1007/s00109-010-0656-8] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Revised: 07/06/2010] [Accepted: 07/08/2010] [Indexed: 01/03/2023]
Abstract
One leading cause of perinatal morbidity and mortality is intrauterine growth restriction (IUGR). Several causes for IUGR have been proposed involving cytotrophoblast dysfunction. Envelope genes of the human endogenous retrovirus (HERV)-W (Syncytin-1), -FRD (Syncytin-2), and -P(b) have fusogenic properties, whereas envelope genes of HERV-R, -V1, and -V2 have putative placental functions. All six HERV envelope genes and three known cellular receptors were analyzed for expression in human control and IUGR placentae (n = 38) and in cultured cytotrophoblasts from control and IUGR (n = 8) placentae. All envelope genes demonstrated downregulation in IUGR compared to control placentae tissues, which were confirmed with cultured cytotrophoblasts. Examination of the Syncytin-1 and Syncytin-2 receptors ASCT-1/-2 and MFSD2 showed that MFSD2 was significantly expressed lower in IUGR than in control placentae and cytotrophoblasts. A reduction of Syncytin-1 protein expression was confirmed for IUGR placentae with immunoblotting and paraffin tissue sections. Embedded placental IUGR tissues showed an overall disorganized syncytiotrophoblast layer with fewer nuclei. Cytotrophoblasts from IUGR placentae demonstrated a lower cell fusion index and nuclei per syncytiotrophoblast in vitro. Fusogenic and non-fusogenic envelope genes are dysregulated in IUGR placentae and may contribute to the etiology of growth restriction in utero.
Collapse
|
41
|
Keating E, Gonçalves P, Costa F, Campos I, Pinho MJ, Azevedo I, Martel F. Comparison of the transport characteristics of bioactive substances in IUGR and normal placentas. Pediatr Res 2009; 66:495-500. [PMID: 19668108 DOI: 10.1203/pdr.0b013e3181b9b4a3] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Knowing that IUGR is associated with altered placental transport, we aimed to characterize the placental transport of folic acid (FA), thiamine (THIAM), serotonin (5-HT), and 1-methyl-4-phenylpyridinium (MPP+) in IUGR. For this, we compared the transport characteristics of (3)H-FA, (3)H-THIAM, (3)H-5-HT, and (3)H-MPP+ in primary cultured human cytotrophoblasts isolated from IUGR and normal placentas (GRTB and NTB cells, respectively) and quantified mRNA expression of several placental transporters, by real-time RT-PCR. Our results show that GRTB cells take up (3)H-FA more efficiently (higher k(in) and A(max) values) and have higher transport capacity (higher V(max) values) for (3)H-FA, (3)H-5-HT, and (3)H-MPP+, when compared with NTB cells. In addition, GRTB cells take up (3)H-THIAM with higher affinity and (3)H-MPP+ with lower affinity than NTB cells. Finally, IUGR placentas have a generalized increase in mRNA expression of FA, THIAM, 5-HT, and MPP+ transporters, when compared with normal placentas, suggesting that the increase in transport capacity may be due to increased expression of placental transporters. These results point to an effect of "compensation for the weakness" of the IUGR placenta and pose the placenta as an active mediator of the communication between maternal and fetal environments.
Collapse
Affiliation(s)
- Elisa Keating
- Department of Biochemistry (U38/FCT), Faculty of Medicine, University of Porto, Porto, Portugal.
| | | | | | | | | | | | | |
Collapse
|
42
|
Melchiorre K, Leslie K, Prefumo F, Bhide A, Thilaganathan B. First-trimester uterine artery Doppler indices in the prediction of small-for-gestational age pregnancy and intrauterine growth restriction. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2009; 33:524-529. [PMID: 19382287 DOI: 10.1002/uog.6368] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
OBJECTIVES To assess the relationship of first-trimester uterine artery Doppler indices with subsequent delivery of small-for-gestational age (SGA) neonates or intrauterine growth restriction (IUGR). METHODS This was a prospective study in which uterine artery Doppler assessment was performed at 11-14 weeks in 3010 women with singleton pregnancies. Resistance indices (RI) and incidence of bilateral notching were compared between women with normal pregnancies (n = 2445) and those who delivered: SGA neonates (birth weight < 10th centile) (n = 377); SGA neonates who had been affected by pre-eclampsia (n = 27); IUGR neonates (birth weight < 10th centile and abnormal Doppler indices) with delivery > or = 37 weeks (n = 62); and IUGR neonates requiring preterm delivery (delivery < 37 weeks) (n = 36). RESULTS The first-trimester uterine artery mean RI and prevalence of bilateral notching were significantly higher in women destined to deliver SGA neonates than in women with normal pregnancies (median uterine artery RI, 0.74 vs. 0.70, P < 0.001; prevalence of bilateral notches, 56% vs. 43%, P < 0.001). The areas under the receiver-operating characteristics curves for the prediction of SGA without pre-eclampsia, IUGR, preterm IUGR and SGA with pre-eclampsia were 0.602, 0.687, 0.776 and 0.708, respectively. There was a statistically significant inverse relationship between mean uterine artery RI and gestational age at delivery in the SGA pregnancies without pre-eclampsia (R = - 0.329, P = 0.01). CONCLUSIONS There is a significant relationship between first-trimester uterine artery Doppler RI and the subsequent development of SGA. The sensitivity of first-trimester uterine artery Doppler is greater for SGA with pre-eclampsia than it is for IUGR alone. This could be because these two entities may have different underlying placental abnormalities that are detected variably on first-trimester uterine artery Doppler evaluation.
Collapse
Affiliation(s)
- K Melchiorre
- Fetal Medicine Unit, Academic Department of Obstetrics and Gynaecology, St George's University of London, London, UK
| | | | | | | | | |
Collapse
|
43
|
Heazell AEP, Crocker IP. Live and let die - regulation of villous trophoblast apoptosis in normal and abnormal pregnancies. Placenta 2008; 29:772-83. [PMID: 18706691 DOI: 10.1016/j.placenta.2008.07.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2008] [Revised: 06/30/2008] [Accepted: 07/03/2008] [Indexed: 01/16/2023]
Abstract
Since 1995 the number of publications investigating apoptosis in villous trophoblast has increased exponentially. This scientific interest is in part due to observations that this specialised form of cell death is increased in pregnancy complications such as pre-eclampsia and intra-uterine growth restriction. In addition, apoptosis is described in normal villous trophoblast and elements of the apoptotic machinery are involved in the fusion between cytotrophoblast and the overlying multinucleate syncytiotrophoblast. The increase in descriptions of apoptotic cell death in villous trophoblast has been accompanied by investigations of regulators of apoptosis. It is anticipated that understanding the regulation of apoptosis in villous trophoblast may provide new insights into placental pathologies. This review describes current knowledge regarding the expression and function of these regulators in villous trophoblast, both in normal and complicated pregnancies.
Collapse
Affiliation(s)
- A E P Heazell
- Maternal and Fetal Health Research Group, St Mary's Hospital, Manchester M13 0JH, UK.
| | | |
Collapse
|
44
|
What matters in preeclampsia are the associated adverse outcomes: the view from Canada. Curr Opin Obstet Gynecol 2008; 20:110-5. [PMID: 18388808 DOI: 10.1097/gco.0b013e3282f733a2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
45
|
Jeyabalan A, McGonigal S, Gilmour C, Hubel C, Rajakumar A. Circulating and placental endoglin concentrations in pregnancies complicated by intrauterine growth restriction and preeclampsia. Placenta 2008; 29:555-63. [PMID: 18462791 PMCID: PMC2467513 DOI: 10.1016/j.placenta.2008.03.006] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2007] [Revised: 03/26/2008] [Accepted: 03/29/2008] [Indexed: 12/20/2022]
Abstract
Inadequate trophoblast invasion and spiral artery remodeling leading to poor placental perfusion and hypoxia are believed to underlie preeclampsia (PE) and intrauterine growth restriction (IUGR). Recent studies implicate increased circulating endoglin as a contributor to the pathogenesis of PE. The objective of this study was to determine whether placental and circulating endoglin concentrations are altered in pregnancies complicated by intrauterine growth restricted (IUGR) infants and to address the role of hypoxia on the regulation of placental endoglin. We analyzed 10 placentas each from normal pregnant (NP), PE, and IUGR subjects. Endoglin levels were 2.5-fold higher in preeclamptic placentas compared to NP (15.4+/-2.6 versus 5.7+/-1.0, p<0.01). In contrast, endoglin levels were similar in NP and IUGR placentas (5.7+/-1.0 vs 5.9+/-1.1, p=NS). Placentas from pregnancies with both PE and IUGR exhibited endoglin levels comparable to the PE group and significantly different from normotensive pregnancies with and without IUGR pregnancies (mean 14.9+/-4.0, n=9, p=0.013). Soluble endoglin concentrations in maternal plasma were comparable in NP and IUGR, but higher in women with PE (n=10 per group, p<0.05). Despite a 2-fold increase in hypoxia inducible factor, HIF-1alpha, we did not observe endoglin upregulation in NP, PE, or IUGR placental villous explants exposed to hypoxia (2% oxygen). In contrast to PE, placental or circulating endoglin is not increased in normotensive women delivering small, asymmetrically grown (IUGR) infants at term. The placentas of women with IUGR appear to be fundamentally different from PE women with respect to endoglin, despite the proposed common pathology of deficient trophoblast invasion/spiral artery remodeling and poor placental perfusion.
Collapse
Affiliation(s)
- A. Jeyabalan
- Department of Obstetrics and Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Magee-Womens Research Institute and Foundation, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - S. McGonigal
- Magee-Womens Research Institute and Foundation, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - C. Gilmour
- Department of Pediatrics, Allegheny General Hospital and Drexel University School of Medicine, Pittsburgh, PA 15212, USA
| | - C.A. Hubel
- Department of Obstetrics and Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Magee-Womens Research Institute and Foundation, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - A. Rajakumar
- Department of Obstetrics and Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Magee-Womens Research Institute and Foundation, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
46
|
Mayhew TM. A stereological perspective on placental morphology in normal and complicated pregnancies. J Anat 2008; 215:77-90. [PMID: 19141109 DOI: 10.1111/j.1469-7580.2008.00994.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Stereology applied to randomly-generated thin sections allows minimally-biased and economical quantitation of the 3D structure of the placenta from molecular to whole-organ levels. With these sampling and estimation tools, it is possible to derive global quantities (tissue volumes, interface surface areas, tubule lengths and particle numbers), average values (e.g. mean cell size or membrane thickness), spatial relationships (e.g. between compartments and immunoprobes) and functional potential (e.g. diffusive conductance). This review indicates ways in which stereology has been used to interpret the morphology of human and murine placentas including the processes of villous growth, trophoblast differentiation, vascular morphogenesis and diffusive transport. In human placenta, global quantities have shown that villous maturation involves differential growth of fetal capillaries and increases in endothelial cell number. Villous trophoblast is a continuously renewing epithelium and, through much of gestation, exhibits a steady state between increasing numbers of nuclei in cytotrophoblast (CT) and syncytiotrophoblast (ST). The epithelium gradually becomes thinner because its surface expands at a faster rate than its volume. These changes help to ensure that placental diffusing capacity matches the growth in fetal mass. Comparable events occur in the murine placenta. Some of these processes are perturbed in complicated pregnancies: 1) fetoplacental vascular growth is compromised in pregnancies accompanied by maternal asthma, 2) changes in trophoblast turnover occur in pre-eclampsia and intrauterine growth restriction, and 3) uteroplacental vascular development is impoverished, but diffusive transport increases, in pregnant mice exposed to particulate urban air pollution. Finally, quantitative immunoelectron microscopy now permits more rigorous analysis of the spatial distributions of interesting molecules between subcellular compartments or shifts in distributions following experimental manipulation.
Collapse
Affiliation(s)
- Terry M Mayhew
- Centre for Integrated Systems Biology & Medicine, School of Biomedical Sciences, Queen's Medical Centre, University of Nottingham, UK.
| |
Collapse
|