1
|
Rossi F, Luppi S, Fejza A, Giolo E, Ricci G, Andreuzzi E. Extracellular matrix and pregnancy: functions and opportunities caught in the net. Reprod Biol Endocrinol 2025; 23:24. [PMID: 39953593 PMCID: PMC11827249 DOI: 10.1186/s12958-025-01348-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 01/24/2025] [Indexed: 02/17/2025] Open
Abstract
The extracellular matrix is a complex network of macromolecules that support the growth and homeostatic development of organisms. By conveying multiple signaling cascades, it impacts on several biological processes and influences the behaviour of numerous cell types. During the endometrial cycle and the key events necessary for a correct embryo implantation and placentation, this bioactive meshwork is substantially modified to favour endometrial receptivity and vascular adaptation, trophoblast cell migration, and immune activation as well. A correct extracellular remodeling is fundamental for the establishment of a physiological pregnancy; indeed, the occurrence of altered matrix modifications associates with gestational disorders such as preeclampsia. In the present review, we will critically evaluate the role of pivotal matrix constituents in regulating the key steps of embryo implantation and placentation, provide up-to-date information concerning their primary mechanisms of action and discuss on their potential as a novel source of biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Francesca Rossi
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, 34137, Italy
| | - Stefania Luppi
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, 34137, Italy
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, 34127, Italy
| | - Albina Fejza
- UBT-Higher Education Institution, Kalabria, Street Rexhep Krasniqi Nr. 56, Prishtina, 10000, Kosovo
| | - Elena Giolo
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, 34137, Italy
| | - Giuseppe Ricci
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, 34137, Italy
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, 34127, Italy
| | - Eva Andreuzzi
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, 34137, Italy.
| |
Collapse
|
2
|
Vidal-Calvo EE, Martin-Salazar A, Choudhary S, Dagil R, Raghavan SSR, Duvnjak L, Nordmaj MA, Clausen TM, Skafte A, Oberkofler J, Wang K, Agerbæk MØ, Løppke C, Jørgensen AM, Ropac D, Mujollari J, Willis S, Garcias López A, Miller RL, Karlsson RTG, Goerdeler F, Chen YH, Colaço AR, Wang Y, Lavstsen T, Martowicz A, Nelepcu I, Marzban M, Oo HZ, Ørum-Madsen MS, Wang Y, Nielsen MA, Clausen H, Wierer M, Wolf D, Gögenur I, Theander TG, Al-Nakouzi N, Gustavsson T, Daugaard M, Salanti A. Tumor-agnostic cancer therapy using antibodies targeting oncofetal chondroitin sulfate. Nat Commun 2024; 15:7553. [PMID: 39215044 PMCID: PMC11364678 DOI: 10.1038/s41467-024-51781-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
Molecular similarities between embryonic and malignant cells can be exploited to target tumors through specific signatures absent in healthy adult tissues. One such embryonic signature tumors express is oncofetal chondroitin sulfate (ofCS), which supports disease progression and dissemination in cancer. Here, we report the identification and characterization of phage display-derived antibody fragments recognizing two distinct ofCS epitopes. These antibody fragments show binding affinity to ofCS in the low nanomolar range across a broad selection of solid tumor types in vitro and in vivo with minimal binding to normal, inflamed, or benign tumor tissues. Anti-ofCS antibody drug conjugates and bispecific immune cell engagers based on these targeting moieties disrupt tumor progression in animal models of human and murine cancers. Thus, anti-ofCS antibody fragments hold promise for the development of broadly effective therapeutic and diagnostic applications targeting human malignancies.
Collapse
Affiliation(s)
- Elena Ethel Vidal-Calvo
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark.
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark.
| | - Anne Martin-Salazar
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Swati Choudhary
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark
| | - Robert Dagil
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark
| | - Sai Sundar Rajan Raghavan
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Lara Duvnjak
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark
| | - Mie Anemone Nordmaj
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Ann Skafte
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark
| | - Jan Oberkofler
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Kaituo Wang
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mette Ø Agerbæk
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- VARCT Diagnostics, Copenhagen, Denmark
| | - Caroline Løppke
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Amalie Mundt Jørgensen
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- VARCT Diagnostics, Copenhagen, Denmark
| | - Daria Ropac
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Joana Mujollari
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Shona Willis
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Agnès Garcias López
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Rebecca Louise Miller
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Richard Torbjörn Gustav Karlsson
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Felix Goerdeler
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Ana R Colaço
- Proteomics Research Infrastructure, University of Copenhagen, Copenhagen, Denmark
| | - Yong Wang
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Thomas Lavstsen
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
| | - Agnieszka Martowicz
- Department of Internal Medicine V, Haematology & Oncology, Comprehensive Cancer Center Innsbruck (CCCI) and Tyrolean Cancer Research Institute (TKFI), Medical University of Innsbruck, Innsbruck, Austria
| | - Irina Nelepcu
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Mona Marzban
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Htoo Zarni Oo
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Maj Sofie Ørum-Madsen
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yuzhuo Wang
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
| | - Morten A Nielsen
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Wierer
- Proteomics Research Infrastructure, University of Copenhagen, Copenhagen, Denmark
| | - Dominik Wolf
- Department of Internal Medicine V, Haematology & Oncology, Comprehensive Cancer Center Innsbruck (CCCI) and Tyrolean Cancer Research Institute (TKFI), Medical University of Innsbruck, Innsbruck, Austria
| | - Ismail Gögenur
- Center for Surgical Science, Department of Surgery, Zealand University Hospital Køge, Køge, Denmark
| | - Thor G Theander
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark
| | - Nader Al-Nakouzi
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Tobias Gustavsson
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark
| | - Mads Daugaard
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark.
- Vancouver Prostate Centre, Vancouver Coastal Health Research Institutes, Vancouver, BC, Canada.
- Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.
| | - Ali Salanti
- Centre for Translational Medicine and Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen University Hospital, Copenhagen, Denmark.
- VAR2 Pharmaceuticals ApS, Copenhagen, Denmark.
| |
Collapse
|
3
|
Slaby EM, Plaisier SB, Brady SR, Hiremath SC, Weaver JD. Controlling placental spheroid growth and phenotype using engineered synthetic hydrogel matrices. Biomater Sci 2024; 12:933-948. [PMID: 38204396 PMCID: PMC10922805 DOI: 10.1039/d3bm01393f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
The human placenta is a complex organ comprised of multiple trophoblast subtypes, and inadequate models to study the human placenta in vitro limit the current understanding of human placental behavior and development. Common in vitro placental models rely on two-dimensional culture of cell lines and primary cells, which do not replicate the native tissue microenvironment, or poorly defined three-dimensional hydrogel matrices such as Matrigel™ that provide limited environmental control and suffer from high batch-to-batch variability. Here, we employ a highly defined, synthetic poly(ethylene glycol)-based hydrogel system with tunable degradability and presentation of extracellular matrix-derived adhesive ligands native to the placenta microenvironment to generate placental spheroids. We evaluate the capacity of a hydrogel library to support the viability, function, and phenotypic protein expression of three human trophoblast cell lines modeling varied trophoblast phenotypes and find that degradable synthetic hydrogels support the greatest degree of placental spheroid viability, proliferation, and function relative to standard Matrigel controls. Finally, we show that trophoblast culture conditions modulate cell functional phenotype as measured by proteomics analysis and functional secretion assays. Engineering precise control of placental spheroid development in vitro may provide an important new tool for the study of early placental behavior and development.
Collapse
Affiliation(s)
- Emily M Slaby
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona, 85287, USA.
| | - Seema B Plaisier
- School of Life Sciences, Arizona State University, Tempe, Arizona, 85287, USA
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona, 85287, USA
| | - Sarah R Brady
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona, 85287, USA.
| | - Shivani C Hiremath
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona, 85287, USA.
| | - Jessica D Weaver
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona, 85287, USA.
| |
Collapse
|
4
|
Antia IU, Hills FA, Shah AJ. Disaccharide compositional analysis of chondroitin sulphate using WAX HILIC-MS with pre-column procainamide labelling; application to the placenta in pre-eclampsia. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:566-575. [PMID: 38189556 DOI: 10.1039/d3ay01578e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Chondroitin sulphate (CS) and dermatan sulphate are negatively charged linear heteropolysaccharides. These glycosaminoglycans (GAG) are involved in cellular signalling via binding to growth factors. CS is expressed in a range of tissue and biological fluids and is highly expressed in the placenta. There is evidence that decorin; a CS proteoglycan is significantly decreased in pre-eclampsia and fetal growth restriction. It is considered that GAG chain composition may influence cellular processes that are altered in pre-eclampsia. The goal of the present study was to develop an LC-MS method with precolumn procainamide labelling for the disaccharide compositional analysis of CS. The method was used to investigate whether the disaccharide composition of placenta-extracted CS is altered in pre-eclampsia. The study revealed differential disaccharide compositions of placental chondroitin sulphate between pre-eclampsia and other pregnancy conditions. This suggests that the method may have diagnostic potential for pregnancy disorders. Furthermore, the findings suggest that CS sulphation might play a significant role in maternal labour.
Collapse
Affiliation(s)
- Imeobong U Antia
- Glycan Research Group, Department of Natural Sciences, Faculty of Science and Technology, Middlesex University, London, UK.
| | - Frank A Hills
- Glycan Research Group, Department of Natural Sciences, Faculty of Science and Technology, Middlesex University, London, UK.
| | - Ajit J Shah
- Glycan Research Group, Department of Natural Sciences, Faculty of Science and Technology, Middlesex University, London, UK.
| |
Collapse
|
5
|
Menkhorst E, So T, Rainczuk K, Barton S, Zhou W, Edgell T, Dimitriadis E. Endometrial stromal cell miR-19b-3p release is reduced during decidualization implying a role in decidual-trophoblast cross-talk. Front Endocrinol (Lausanne) 2023; 14:1149786. [PMID: 37008948 PMCID: PMC10061138 DOI: 10.3389/fendo.2023.1149786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/03/2023] [Indexed: 03/18/2023] Open
Abstract
INTRODUCTION A healthy pregnancy requires successful blastocyst implantation into an adequately prepared or 'receptive' endometrium. Decidualization of uterine endometrial stromal fibroblast cells (hESF) is critical for the establishment of a healthy pregnancy. microRNAs (miRs) are critical regulators of cellular function that can be released by a donor cell to influence the physiological state of recipient cells. We aimed to determine how decidualization affects hESF miR release and investigated the function of one decidualization regulated miR, miR-19b-3p, previously shown to be associated with recurrent pregnancy loss. METHOD miR release by hESF was determined by miR microarray on culture media from hESF decidualized in vitro for 3 and 14 days by treatment with oestradiol and medroxyprogesterone acetate. Cellular and whole endometrial/decidual tissue miR expression was quantified by qPCR and localized by in situ hybridization. The function of miR-19b-3p in HTR8/Svneo trophoblast cells was investigated using real time cell analysis (xCELLigence) and gene expression qPCR. RESULTS From our miR screen we found that essentially all hESF miR release was reduced following in vitro decidualization, significantly so for miR-17-5p, miR-21-3p, miR-34c-3p, miR-106b-5p, miR-138-5p, miR-296-5p, miR-323a-3p, miR-342-3p, miR-491-5p, miR-503-5p and miR-542-5p. qPCR demonstrated that miR-19b-3p, 181a-2-3p and miR-409-5p likewise showed a significant reduction in culture media following decidualization but no change was found in cellular miR expression following decidualization. In situ hybridization localized miR-19b-3p to epithelial and stromal cells in the endometrium and qPCR identified that miR-19b-3p was significantly elevated in the cycling endometrium of patients with a history of early pregnancy loss compared to normally fertile controls. Functionally, overexpression of miR-19b-3p significantly reduced HTR8/Svneo trophoblast proliferation and increased HOXA9 expression. DISCUSSION Our data demonstrates that decidualization represses miR release by hESFs and overexpression of miR-19b-3p was found in endometrial tissue from patients with a history of early pregnancy loss. miR-19b-3p impaired HTR8/Svneo proliferation implying a role in trophoblast function. Overall we speculate that miR release by hESF may regulate other cell types within the decidua and that appropriate release of miRs by decidualized hESF is essential for healthy implantation and placentation.
Collapse
Affiliation(s)
- Ellen Menkhorst
- Embryo Implantation Laboratory, Department of Obstetrics and Gynecology, The University of Melbourne, Parkville, VIC, Australia
- Gynecology Research Centre, The Royal Women’s Hospital, Parkville, VIC, Australia
- *Correspondence: Ellen Menkhorst,
| | - Teresa So
- Embryo Implantation Laboratory, Department of Obstetrics and Gynecology, The University of Melbourne, Parkville, VIC, Australia
- Gynecology Research Centre, The Royal Women’s Hospital, Parkville, VIC, Australia
| | - Kate Rainczuk
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Siena Barton
- Embryo Implantation Laboratory, Department of Obstetrics and Gynecology, The University of Melbourne, Parkville, VIC, Australia
- Gynecology Research Centre, The Royal Women’s Hospital, Parkville, VIC, Australia
| | - Wei Zhou
- Embryo Implantation Laboratory, Department of Obstetrics and Gynecology, The University of Melbourne, Parkville, VIC, Australia
- Gynecology Research Centre, The Royal Women’s Hospital, Parkville, VIC, Australia
| | - Tracey Edgell
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Evdokia Dimitriadis
- Embryo Implantation Laboratory, Department of Obstetrics and Gynecology, The University of Melbourne, Parkville, VIC, Australia
- Gynecology Research Centre, The Royal Women’s Hospital, Parkville, VIC, Australia
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
| |
Collapse
|
6
|
Decorin Promotes Osteoblastic Differentiation of Human Periodontal Ligament Stem Cells. Molecules 2022; 27:molecules27238224. [PMID: 36500314 PMCID: PMC9739490 DOI: 10.3390/molecules27238224] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/14/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
The aim of this study is to clarify the biological functions of decorin (DCN) in the healing and regeneration of wounded periodontal tissue. We investigated the expression pattern of DCN during the healing of wounded periodontal tissue in rats by immunohistochemistry and the effects of DCN on the osteoblastic differentiation of human periodontal ligament (PDL) stem cells (HPDLSCs) and preosteoblasts by Alizarin red S staining, quantitative reverse transcription-polymerase chain reactions, and western blotting. The expression of DCN was increased around the wounded PDL tissue on day 5 after surgery compared with the nonwounded PDL tissue, whereas its expression was not changed in the osteoblastic layer around the wounded alveolar bone. Furthermore, DCN promoted the osteoblastic differentiation of HPDLSCs, but it did not affect the osteoblastic differentiation of preosteoblasts. ERK1/2 phosphorylation was upregulated during the DCN-induced osteoblastic differentiation of HPDLSCs. DCN did not affect proliferation, migration, or the PDL-related gene expression of HPDLSCs. In conclusion, this study demonstrates that DCN has a role in the healing of wounded periodontal tissue. Furthermore, DCN secreted from PDL cells may contribute to bone healing by upregulating osteoblastic differentiation through ERK1/2 signaling in HPDLSCs, indicating a therapeutic effect of DCN in periodontal tissue regeneration.
Collapse
|
7
|
Halari CD, Nandi P, Sidhu J, Sbirnac M, Zheng M, Lala PK. Decorin–induced, preeclampsia-associated microRNA-512-3p restrains extravillous trophoblast functions by targeting USF2/PPP3R1 axis. Front Cell Dev Biol 2022; 10:1014672. [PMID: 36299488 PMCID: PMC9588925 DOI: 10.3389/fcell.2022.1014672] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/22/2022] [Indexed: 11/24/2022] Open
Abstract
Decorin (DCN) is a leucine-rich proteoglycan produced by chorionic villus mesenchymal cells anddecidual cells during human pregnancy. Studies from our laboratory demonstrated that decidua-derived DCN restrains multiple trophoblast functions including proliferation, migration, invasion andendovascular differentiation, mediated by DCN-binding to multiple tyrosine kinase receptors; expressed by the trophoblast. Furthermore, DCN was shown to be selectively over-produced by thedecidua in preeclampsia (PE) subjects and elevated in the second trimester maternal plasma in PE, before the appearance of clinical signs, presenting as a predictive biomarker for PE. Micro (mi)RNAs are single-stranded non-coding RNAs (17–25 nucleotides) that typically downregulate target genes by repressing translation or facilitating degradation of mRNAs. The human; placenta expresses many miRNAs, some of which are exclusively expressed by the trophoblast. Many; of these miRNAs are dysregulated in PE-associated placentas and some appear in the maternal blood as PE biomarkers. However, little is known about their contribution to the pathogenesis of PE, a multi-factorial disease associated with a hypo-invasive placenta. The objective of the present study was to examine whether exposure of extravillous trophoblast (EVT) to DCN affects expression of specific miRNAs, and to test the role of these miRNAs in altering EVT functions. We identified miR-512-3p, as one of the DCN-induced miRNAs, also upregulated in PE placentas. It was shown to be elevated in ectopic DCN-over-expressing or exogenous DCN-treated first trimester human trophoblast cell line HTR-8/SVneo. Use of miRNA-mimics and inhibitors revealed that miR-512-3p compromised trophoblast migration, invasion and VEGF-dependent endovascular differentiation. Finally, Protein Phosphatase 3 Regulatory Subunit B, Alpha (PPP3R1), a known target of miR-512-3p, was paradoxically elevated in miR-512-3p-overexpressing trophoblast and PE-associated placentas. Using Enrichr, a tool that consists of both a validated user-submitted gene list and a search engine for transcription factors, we found that PPP3R1 elevation resulted from the miRNA binding to and targeting Upstream Transcription Factor 2 (USF2) which targeted PPP3R1. These findings reveal a novel aspect of pathogenesis of PE and biomarker potentials of this miRNA in PE.
Collapse
Affiliation(s)
- Chidambra D. Halari
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Pinki Nandi
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Jasmin Sidhu
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Maria Sbirnac
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Michael Zheng
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Peeyush K. Lala
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
- Children’s Health Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
- *Correspondence: Peeyush K. Lala,
| |
Collapse
|
8
|
Hayder H, Shan Y, Chen Y, O’Brien JA, Peng C. Role of microRNAs in trophoblast invasion and spiral artery remodeling: Implications for preeclampsia. Front Cell Dev Biol 2022; 10:995462. [PMID: 36263015 PMCID: PMC9575991 DOI: 10.3389/fcell.2022.995462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/25/2022] [Indexed: 11/18/2022] Open
Abstract
It is now well-established that microRNAs (miRNAs) are important regulators of gene expression. The role of miRNAs in placental development and trophoblast function is constantly expanding. Trophoblast invasion and their ability to remodel uterine spiral arteries are essential for proper placental development and successful pregnancy outcome. Many miRNAs are reported to be dysregulated in pregnancy complications, especially preeclampsia and they exert various regulatory effects on trophoblasts. In this review, we provide a brief overview of miRNA biogenesis and their mechanism of action, as well as of trophoblasts differentiation, invasion and spiral artery remodeling. We then discuss the role of miRNAs in trophoblasts invasion and spiral artery remodeling, focusing on miRNAs that have been thoroughly investigated, especially using multiple model systems. We also discuss the potential role of miRNAs in the pathogenesis of preeclampsia.
Collapse
Affiliation(s)
- Heyam Hayder
- Department of Biology, York University, Toronto, ON, Canada
| | - Yanan Shan
- Department of Biology, York University, Toronto, ON, Canada
| | - Yan Chen
- Department of Biology, York University, Toronto, ON, Canada
| | | | - Chun Peng
- Department of Biology, York University, Toronto, ON, Canada
- Centre for Research on Biomolecular Interactions, York University, Toronto, ON, Canada
- *Correspondence: Chun Peng,
| |
Collapse
|
9
|
Vijayan AN, Solaimuthu A, Murali P, Gopi J, Y MT, R AP, Korrapati PS. Decorin mediated biomimetic PCL-gelatin nano-framework to impede scarring. Int J Biol Macromol 2022; 219:907-918. [PMID: 35952816 DOI: 10.1016/j.ijbiomac.2022.08.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/03/2022] [Accepted: 08/06/2022] [Indexed: 11/05/2022]
Abstract
Scars occur as a result of fibrosis after tissue damage or surgery and reports suggest that excessive Transforming growth factor-β (TGF-β) activity during the process of wound healing leads to progressive fibrosis. Decorin is an extracellular matrix (ECM) protein which regulates collagen fibrillogenesis. However, targeted delivery and effective protein therapy remains a challenge owing to degradation byproteases. Hence, we aimed to deliver Decorin in a sustainable mode for the reduction of TGF-β levels and subsequent scar formation. Herein, we have fabricated PCL-Gelatin bio-mimetic scaffolds to optimize the bio-activity and provide localized delivery of recombinant Decorin. The degradation and drug release patterns reveals that this biomaterial is biodegradable and offers sustained release of the recombinant Decorin. Decorin loaded nanofiber displayed lower adhesion and proliferation rates in in-vitro conditions. Moreover, Decorin loaded scaffolds demonstrated morphological changes in cells, specifically targeting the myofibroblast. The expression of TGF-β was also scrutinized to understand the effect of Decorin loaded nanofibers. Besides, in the in-vitro fibrotic model, Decorin loaded nanofibers efficiently reduced the expression of ECM related proteins. Therefore, we report the sustained delivery of the recombinant Decorin from nanofiber dressing to potentially obstruct scar formation during the process of wound healing.
Collapse
Affiliation(s)
- Ane Nishitha Vijayan
- Biological Materials Laboratory, CSIR-Central Leather Research Institute (CSIR-CLRI), Adyar, Chennai 600020, Tamil Nadu, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Anbuthiruselvan Solaimuthu
- Biological Materials Laboratory, CSIR-Central Leather Research Institute (CSIR-CLRI), Adyar, Chennai 600020, Tamil Nadu, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Padmaja Murali
- Biological Materials Laboratory, CSIR-Central Leather Research Institute (CSIR-CLRI), Adyar, Chennai 600020, Tamil Nadu, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Janani Gopi
- Biological Materials Laboratory, CSIR-Central Leather Research Institute (CSIR-CLRI), Adyar, Chennai 600020, Tamil Nadu, India
| | - Madhan Teja Y
- Biological Materials Laboratory, CSIR-Central Leather Research Institute (CSIR-CLRI), Adyar, Chennai 600020, Tamil Nadu, India
| | - Akshaya Priya R
- Biological Materials Laboratory, CSIR-Central Leather Research Institute (CSIR-CLRI), Adyar, Chennai 600020, Tamil Nadu, India
| | - Purna Sai Korrapati
- Biological Materials Laboratory, CSIR-Central Leather Research Institute (CSIR-CLRI), Adyar, Chennai 600020, Tamil Nadu, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
10
|
Oravecz O, Balogh A, Romero R, Xu Y, Juhasz K, Gelencser Z, Xu Z, Bhatti G, Pique-Regi R, Peterfia B, Hupuczi P, Kovalszky I, Murthi P, Tarca AL, Papp Z, Matko J, Than NG. Proteoglycans: Systems-Level Insight into Their Expression in Healthy and Diseased Placentas. Int J Mol Sci 2022; 23:5798. [PMID: 35628608 PMCID: PMC9147780 DOI: 10.3390/ijms23105798] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/14/2022] [Accepted: 05/15/2022] [Indexed: 02/04/2023] Open
Abstract
Proteoglycan macromolecules play key roles in several physiological processes (e.g., adhesion, proliferation, migration, invasion, angiogenesis, and apoptosis), all of which are important for placentation and healthy pregnancy. However, their precise roles in human reproduction have not been clarified. To fill this gap, herein, we provide an overview of the proteoglycans' expression and role in the placenta, in trophoblast development, and in pregnancy complications (pre-eclampsia, fetal growth restriction), highlighting one of the most important members of this family, syndecan-1 (SDC1). Microarray data analysis showed that of 34 placentally expressed proteoglycans, SDC1 production is markedly the highest in the placenta and that SDC1 is the most upregulated gene during trophoblast differentiation into the syncytiotrophoblast. Furthermore, placental transcriptomic data identified dysregulated proteoglycan genes in pre-eclampsia and in fetal growth restriction, including SDC1, which is supported by the lower concentration of syndecan-1 in maternal blood in these syndromes. Overall, our clinical and in vitro studies, data analyses, and literature search pointed out that proteoglycans, as important components of the placenta, may regulate various stages of placental development and participate in the maintenance of a healthy pregnancy. Moreover, syndecan-1 may serve as a useful marker of syncytialization and a prognostic marker of adverse pregnancy outcomes. Further studies are warranted to explore the role of proteoglycans in healthy and complicated pregnancies, which may help in diagnostic or therapeutic developments.
Collapse
Affiliation(s)
- Orsolya Oravecz
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (O.O.); (A.B.); (K.J.); (Zs.G.); (B.P.); (J.M.)
- Doctoral School of Biology, Institute of Biology, ELTE Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Andrea Balogh
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (O.O.); (A.B.); (K.J.); (Zs.G.); (B.P.); (J.M.)
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (Z.X.); (G.B.); (R.P.-R.); (A.L.T.)
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
- Detroit Medical Center, Detroit, MI 48201, USA
| | - Yi Xu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (Z.X.); (G.B.); (R.P.-R.); (A.L.T.)
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
| | - Kata Juhasz
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (O.O.); (A.B.); (K.J.); (Zs.G.); (B.P.); (J.M.)
| | - Zsolt Gelencser
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (O.O.); (A.B.); (K.J.); (Zs.G.); (B.P.); (J.M.)
| | - Zhonghui Xu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (Z.X.); (G.B.); (R.P.-R.); (A.L.T.)
| | - Gaurav Bhatti
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (Z.X.); (G.B.); (R.P.-R.); (A.L.T.)
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
| | - Roger Pique-Regi
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (Z.X.); (G.B.); (R.P.-R.); (A.L.T.)
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
| | - Balint Peterfia
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (O.O.); (A.B.); (K.J.); (Zs.G.); (B.P.); (J.M.)
| | | | - Ilona Kovalszky
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary;
| | - Padma Murthi
- Department of Pharmacology, Monash Biomedicine Discovery Institute, Clayton, VIC 3800, Australia;
- Department of Obstetrics and Gynaecology, University of Melbourne, Royal Women’s Hospital, Parkville, VIC 3502, Australia
| | - Adi L. Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892, and Detroit, MI 48201, USA; (R.R.); (Y.X.); (Z.X.); (G.B.); (R.P.-R.); (A.L.T.)
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI 48202, USA
| | - Zoltan Papp
- Maternity Private Clinic, H-1126 Budapest, Hungary; (P.H.); (Z.P.)
| | - Janos Matko
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (O.O.); (A.B.); (K.J.); (Zs.G.); (B.P.); (J.M.)
| | - Nandor Gabor Than
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, H-1117 Budapest, Hungary; (O.O.); (A.B.); (K.J.); (Zs.G.); (B.P.); (J.M.)
- Maternity Private Clinic, H-1126 Budapest, Hungary; (P.H.); (Z.P.)
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, H-1085 Budapest, Hungary;
| |
Collapse
|
11
|
Park BS, Lee J, Jun JH. Decorin: a multifunctional proteoglycan involved in oocyte maturation and trophoblast migration. Clin Exp Reprod Med 2021; 48:303-310. [PMID: 34875737 PMCID: PMC8651757 DOI: 10.5653/cerm.2021.05071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 11/15/2021] [Indexed: 12/02/2022] Open
Abstract
Decorin (DCN) is a proteoglycan belonging to the small leucine-rich proteoglycan family. It is composed of a protein core containing leucine repeats with a glycosaminoglycan chain consisting of either chondroitin sulfate or dermatan sulfate. DCN is a structural component of connective tissues that can bind to type I collagen. It plays a role in the assembly of the extracellular matrix (ECM), and it is related to fibrillogenesis. It can interact with fibronectin, thrombospondin, complement component C1, transforming growth factor (TGF), and epidermal growth factor receptor. Normal DCN expression regulates a wide range of cellular processes, including proliferation, migration, apoptosis, and autophagy, through interactions with various molecules. However, its aberrant expression is associated with oocyte maturation, oocyte quality, and poor extravillous trophoblast invasion of the uterus, which underlies the occurrence of preeclampsia and intrauterine growth restriction. Spatiotemporal hormonal control of successful pregnancy should regulate the concentration and activity of specific proteins such as proteoglycan participating in the ECM remodeling of trophoblastic and uterine cells in fetal membranes and uterus. At the human feto-maternal interface, TGF-β and DCN play crucial roles in the regulation of trophoblast invasion of the uterus. This review summarizes the role of the proteoglycan DCN as an important and multifunctional molecule in the physiological regulation of oocyte maturation and trophoblast migration. This review also shows that recombinant DCN proteins might be useful for substantiating diverse functions in both animal and in vitro models of oogenesis and implantation.
Collapse
Affiliation(s)
- Beom Seok Park
- Department of Biomedical Laboratory Science, College of Health Science, Eulji University, Seongnam, Korea.,Department of Biomedical Laboratory Science, Graduate School, Eulji University, Seongnam, Korea.,Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Seongnam, Korea
| | - Jaewang Lee
- Department of Biomedical Laboratory Science, College of Health Science, Eulji University, Seongnam, Korea.,Department of Biomedical Laboratory Science, Graduate School, Eulji University, Seongnam, Korea
| | - Jin Hyun Jun
- Department of Biomedical Laboratory Science, College of Health Science, Eulji University, Seongnam, Korea.,Department of Biomedical Laboratory Science, Graduate School, Eulji University, Seongnam, Korea.,Department of Senior Healthcare, BK21 Plus Program, Graduate School, Eulji University, Seongnam, Korea.,Eulji Medi-Bio Research Institute (EMBRI), Eulji University, Daejeon, Korea
| |
Collapse
|
12
|
Halari CD, Renaud SJ, Lala PK. Molecular mechanisms in IL-1β-mediated decorin production by decidual cells. Mol Hum Reprod 2021; 27:6463501. [PMID: 34915564 DOI: 10.1093/molehr/gaab068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/06/2021] [Indexed: 12/15/2022] Open
Abstract
Decorin, a small leucine-rich proteoglycan produced by decidual cells restrains trophoblast differentiation, migration and invasiveness of extra-villous trophoblast cells. Decidual overproduction of decorin is associated with preeclampsia, and elevated decorin levels in maternal plasma are a predictive biomarker of preeclampsia. Furthermore, decorin plays an autocrine role in maturation of human endometrial stromal cells into decidual cells. Thus, a balanced decorin production by the decidua is critical for healthy pregnancy. However, the molecular mechanisms regulating decorin production by the decidua are unclear. Interleukin-1 beta is an inflammation-associated multi-functional cytokine, and is reported to induce decidualization in primates. Hence, the present study was designed: (i) to test if exogenous Interleukin-1 beta stimulated decorin production by human endometrial stromal cells; and if so, (ii) to identify the cellular source of Interleukin-1 beta in first trimester decidual tissue; (iii) to identify the downstream molecular partners in Interleukin-1 beta mediated decorin production by human endometrial stromal cells. Results revealed that (i) amongst multiple pro-inflammatory cytokines tested, Interleukin-1 beta alone stimulated decorin production by these cells; (ii) both macrophages and decidual cells in first trimester decidua produced Interleukin-1 beta; (iii) Interleukin-1 beta mediated decorin production was dependent on Interleukin-1 receptor activation, followed by activation and nuclear translocation of nuclear factor kappa B and its binding to the decorin promoter. These results reveal that Interleukin-1 beta plays a novel role in inducing decorin production by human endometrial stromal cells by activating nuclear factor kappa B.
Collapse
Affiliation(s)
- C D Halari
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
| | - S J Renaud
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada.,Children's Health Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
| | - P K Lala
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada.,Children's Health Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada.,Department of Oncology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
| |
Collapse
|
13
|
Roles of Two Small Leucine-Rich Proteoglycans Decorin and Biglycan in Pregnancy and Pregnancy-Associated Diseases. Int J Mol Sci 2021; 22:ijms221910584. [PMID: 34638928 PMCID: PMC8509074 DOI: 10.3390/ijms221910584] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 11/18/2022] Open
Abstract
Two small leucine-rich proteoglycans (SLRP), decorin and biglycan, play important roles in structural–functional integrity of the placenta and fetal membranes, and their alterations can result in several pregnancy-associated diseases. In this review, we briefly discuss normal placental structure and functions, define and classify SLRPs, and then focus on two SLRPs, decorin (DCN) and biglycan (BGN). We discuss the consequences of deletions/mutations of DCN and BGN. We then summarize DCN and BGN expression in the pregnant uterus, myometrium, decidua, placenta, and fetal membranes. Actions of these SLRPs as ligands are then discussed in the context of multiple binding partners in the extracellular matrix and cell surface (receptors), as well as their alterations in pathological pregnancies, such as preeclampsia, fetal growth restriction, and preterm premature rupture of membranes. Lastly, we raise some unanswered questions as food for thought.
Collapse
|
14
|
Halari CD, Nandi P, Jeyarajah MJ, Renaud SJ, Lala PK. Decorin production by the human decidua: role in decidual cell maturation. Mol Hum Reprod 2021; 26:784-796. [PMID: 32866233 DOI: 10.1093/molehr/gaaa058] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 08/11/2020] [Indexed: 01/13/2023] Open
Abstract
Decidualization involves the proliferation and differentiation of fibroblast-like endometrial stromal cells into epithelioid-shaped and secretory 'decidual' cells in response to steroid hormones. Human decidual cells produce insulin-like growth factor-binding protein-1 and prolactin (PRL), two well-recognized markers of decidual cell maturation and a proteoglycan decorin (DCN). We reported that DCN restrains the human trophoblast renewal, migration, invasion and endovascular differentiation needed for uterine arterial remodeling during normal pregnancy. DCN overproduction by the decidua is associated with a hypo-invasive placenta and a serious pregnancy disorder, pre-eclampsia (PE). Furthermore, elevated maternal plasma DCN levels during the second trimester is a predictive biomarker of PE. While these paracrine roles of decidua-derived DCN on trophoblast physiology and pathology have been well-defined, it remains unknown whether DCN plays any autocrine role in decidual cell development. The objectives of this study were to examine: the kinetics of DCN production during decidualization of human endometrial stromal cells; gestational age-related changes in DCN production by the first trimester decidua; and a possible autocrine role of DCN on decidual cell maturation. We found that DCN production is enhanced during decidualization of both primary and immortalized human endometrial stromal cells in vitro and during early gestation in decidual samples tested ex vivo, and that it is important for endometrial stromal cell maturation into a decidual phenotype. Decorin-depleted human endometrial stromal cells exposed to decidualizing stimuli failed to mature fully, as evidenced by fibroblastoid morphology, reduced insulin-like growth factor-binding protein-1 and PRL expression, and reduction in cellular ploidy. We identified heart and neural crest derivatives-expressed protein 2, and progesterone receptor as potential downstream mediators of DCN effects.
Collapse
Affiliation(s)
- C D Halari
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
| | - P Nandi
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
| | - M J Jeyarajah
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
| | - S J Renaud
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada.,Children's Health Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
| | - P K Lala
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada.,Children's Health Research Institute, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada.,Department of Oncology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
| |
Collapse
|
15
|
Lu Y, Li X, Zuo Y, Xu Q, Liu L, Wu H, Chen L, Zhang Y, Liu Y, Li Y. miR-373-3p inhibits epithelial-mesenchymal transition via regulation of TGFβR2 in choriocarcinoma. J Obstet Gynaecol Res 2021; 47:2417-2432. [PMID: 33955122 DOI: 10.1111/jog.14809] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 02/24/2021] [Accepted: 04/18/2021] [Indexed: 12/19/2022]
Abstract
AIM Previous studies have indicated that early metastasis is a major cause of mortality in patients with choriocarcinoma. However, what determines whether early metastasis of choriocarcinoma has occurred is unknown. The emerging role of miRNA in regulating cancer development and progression has been recognized. miR-373 has been shown to play pivotal roles in tumorigenesis and metastasis. However, whether miR-373 functions to promote choriocarcinoma metastasis is not clear. The purpose of this study is to determine the function of miR-373-3p in the progression of this cancer. METHODS In this study, we first compared epithelial-mesenchymal transition (EMT)-related markers, which were inversely correlated with miR-373-3p expression in trophoblast and choriocarcinoma cell lines. Using PCR and Western blot, upregulation of miR-373-3p was observed to inhibit EMT progression. Similarly, gain- and loss-of-function studies revealed that ectopic miR-373-3p overexpression inhibited the migration by transwell methods of choriocarcinoma cells. RESULTS Our results revealed that miR-373-3p acted as an EMT inhibitor in JEG-3 and JAR cells; this was due to its mediation of the transforming growth factor-β (TGFβ) signaling pathway, which was responsible for EMT. miRNA microarray analysis demonstrated that miR-373-3p interacted with the 3' untranslated region of TGFβR2 mRNA, and then Western blot and dual-luciferase reporter gene assays verified this interaction. CONCLUSION Our findings suggest that miR-373-3p upregulation partly accounts for TGFβR2 downregulation and leads to a restraint of EMT and migration. miR-373-3p may therefore serve as a valuable potential target in the treatment of choriocarcinoma.
Collapse
Affiliation(s)
- Yanjie Lu
- Department of Pathology, Chengde Medical University, Chengde, Hebei, China.,Cancer Research Laboratory, Chengde Medical University, Chengde, Hebei, China
| | - Xiaoru Li
- Cancer Research Laboratory, Chengde Medical University, Chengde, Hebei, China.,Department of Gynaecology and Obstetrics, Chengde Central Hospital, Chengde, Hebei, China
| | - Yanzhen Zuo
- Cancer Research Laboratory, Chengde Medical University, Chengde, Hebei, China
| | - Qian Xu
- Cancer Research Laboratory, Chengde Medical University, Chengde, Hebei, China
| | - Lei Liu
- Cancer Research Laboratory, Chengde Medical University, Chengde, Hebei, China
| | - Haiying Wu
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Chengde Medical University, Chengde, Hebei, China
| | - Long Chen
- Department of Pathology, Chengde Medical University, Chengde, Hebei, China
| | - Ying Zhang
- Department of Pathology, Chengde Medical University, Chengde, Hebei, China
| | - Ying Liu
- Department of Pathology, Chengde Medical University, Chengde, Hebei, China
| | - Yuhong Li
- Department of Pathology, Chengde Medical University, Chengde, Hebei, China.,Cancer Research Laboratory, Chengde Medical University, Chengde, Hebei, China
| |
Collapse
|
16
|
Lala PK, Nandi P, Hadi A, Halari C. A crossroad between placental and tumor biology: What have we learnt? Placenta 2021; 116:12-30. [PMID: 33958236 DOI: 10.1016/j.placenta.2021.03.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 02/27/2021] [Accepted: 03/02/2021] [Indexed: 01/06/2023]
Abstract
Placenta in certain species including the human has evolved as a highly invasive tumor-like organ invading the uterus aned its vasculature to derive oxygen and nutrients for the fetus and exchange waste products. While several excellent reviews have been written comparing hemochorial placentation with tumors, no comprehensive review is available dealing with mechanistic insights into what makes them different, and what tumor biologists can learn from placental biologists, and vice versa. In this review, we analyze the structure-function relationship of the human placenta, emphasizing the functional need of the spatio-temporally orchestrated trophoblast invasiveness for fetal development and growth, and pathological consequences of aberrant invasiveness for fetal and maternal health. We then analyze similarities and differences between the placenta and invasive tumors in terms of hallmarks of cancer, some key molecules regulating their invasive functions, and how placental cancers (choriocarcinomas) or other cancers become refractory or even addicted to these invasion-restraining molecules. We cite in vitro models of human trophoblast and choriocarcinoma cell lines utilized to study mechanisms in normal placental development as well as those responsible for tumor progression. We discuss the pathobiology of hyper-invasive placentas and show thattrophoblastic neoplasias are a unique and heterogeneous class of tumors. We delve into the questions as to why metastasis from other organs rarely occurs at the placental site and whether pregnancy makes the mother more or less vulnerable to cancer-related morbidity/mortality. We attempt to compare trophoblast stem cells and cancer stem cells. Finally, we leave the readers with some thoughts as foods of future investigations.
Collapse
Affiliation(s)
- Peeyush K Lala
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Westernat Ontario, London, Ontario, N6A5C1, Canada; Associate Scientist, Children's Health Research Institute, University of Western Ontario, London, Ontario, Canada N6C2V5.
| | - Pinki Nandi
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Westernat Ontario, London, Ontario, N6A5C1, Canada.
| | - Ali Hadi
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Westernat Ontario, London, Ontario, N6A5C1, Canada.
| | - Chidambra Halari
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Westernat Ontario, London, Ontario, N6A5C1, Canada.
| |
Collapse
|
17
|
Adanaş Aydın G, Ayvacı H, Özgen G. The first-trimester serum decorin levels as a potential predictor of preeclampsia. J Perinat Med 2020; 48:779-785. [PMID: 32739907 DOI: 10.1515/jpm-2020-0178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 06/24/2020] [Indexed: 11/15/2022]
Abstract
Background Preeclampsia (PE) is a multisystem disease and is still among the leading causes of maternal and neonatal morbidity and mortality. Inadequate trophoblast invasion plays a key role in the PE pathogenesis. The proliferation, migration, and invasion of extravillous trophoblasts (EVTs) is primarily controlled by the decidua-derived transforming growth factor beta (TGF-β) and decorin. In this study, we aimed to investigate the clinical utility of serum decorin levels measured in the 11th to 14th gestational weeks to predict preeclampsia during the following weeks of gestation. Materials and Methods A total of 600 pregnant women, whose age and gestational age ranged from 18 to 40 years and 11 to 14 weeks, were included. Venous blood samples were obtained and stored at -80 °C. Subsequently, the patients who developed preeclampsia and healthy controls with a similar body mass index were identified and their first-trimester blood samples were analyzed for serum decorin levels. Results The mean serum decorin level was 8.76 ± 6.88 ng/mL for the PE group while 9.75 ± 9.82 ng/mL for the control group. No statistically significant difference was found between the two groups (p=0.838). Conclusion We observed that the serum decorin levels during the 11th to 14th weeks of gestation showed no predictive value for preeclampsia in pregnant women. However, more accurate conclusions about the clinical utility of decorin as a biomarker of preeclampsia would require further studies with larger samples including more patients with EOS-PE.
Collapse
Affiliation(s)
- Gültekin Adanaş Aydın
- Department of Obstetrics and Gynecology, Bursa Yüksek İhtisas Training and Research Hospital, Bursa, Turkey
| | - Habibe Ayvacı
- Department of Obstetrics and Gynecology, Zeynep Kamil Women's and Children's Disease Training and Research Hospital, Istanbul, Turkey
| | - Gülten Özgen
- Department of Obstetrics and Gynecology, Bursa Yüksek İhtisas Training and Research Hospital, Bursa, Turkey
| |
Collapse
|
18
|
Borovkov VA, Igitova MB, Korenovskiy YV, Dudareva YA. [Prognostic significance of specific proteins of pregnancy in women with a uterine scar and placenta accreta.]. Klin Lab Diagn 2020; 65:353-357. [PMID: 32459893 DOI: 10.18821/0869-2084-2020-65-6-353-357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 04/10/2020] [Indexed: 11/17/2022]
Abstract
Comparative analysis of serum concentrations of chorionic gonadotropin (hCG) associated with the pregnancy of plasma protein A (PAPP-A) and alpha-fetoprotein (AFP), based on the results of a survey of women as part of a standard screening program (the results were expressed as a MoM - multiply of the median), found a significant increase in the performance of all the studied specific pregnancy proteins in women with a scar on the uterus and placenta acctera (75 patients) compared with the data of the group of pregnant women without scar on the uterus and without abnormalities of attachment of the placenta (150 women). AFP indices were 1.68 ± 0.76 and 1.19 ± 0.43 MoM (p = 0.0018), hCG - 1.62 ± 1.48 and 1.23 ± 0.76 MoM (p = 0, 0112), PAPP-A - 1.93 ± 1.24 and 1.23 ± 0.67 MoM (p <0.0001). Using the ROC analysis, the diagnostic thresholds for the concentrations of AFP, hCG and PAPP-A were calculated. The risk of placenta accreta in women with a scar on the uterus in cases of exceeding the diagnostic threshold of AFP concentration (1.64 MoM) increased 2.5 times (RR = 2.5; 95% CI 1.17-5.36, p = 0, 0185), hCG (1.41 MoM) - 1.6 times (RR = 1.59; 95% CI 1.09-2.32, p = 0.0147), PAPP-A (1.41 MoM) - 2.65 times (RR = 2.65; 95% CI 1.76-3.99, p <0.0001). Determination of the level of specific pregnancy proteins can be used in the system of complex prediction of placental growth in pregnant women with a scar on the uterus as an addition to the assessment of clinical and anamnestic risk factors.
Collapse
Affiliation(s)
- V A Borovkov
- Altai Regional Clinical Center for Maternity and Child Welfare Surveillance, Barnaul, Russian Federation
| | - M B Igitova
- Altai State Medical University, Barnaul, Russian Federation
| | | | - Y A Dudareva
- Altai State Medical University, Barnaul, Russian Federation
| |
Collapse
|
19
|
Three types of HLA-G+ extravillous trophoblasts that have distinct immune regulatory properties. Proc Natl Acad Sci U S A 2020; 117:15772-15777. [PMID: 32581122 DOI: 10.1073/pnas.2000484117] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
During pregnancy, invading HLA-G+ extravillous trophoblasts (EVT) play a key role in placental development, uterine spiral artery remodeling, and prevention of detrimental maternal immune responses to placental and fetal antigens. Failures of these processes are suggested to play a role in the development of pregnancy complications, but very little is known about the underlying mechanisms. Here we present validated methods to purify and culture primary HLA-G+ EVT from the placental disk and chorionic membrane from healthy term pregnancy. Characterization of HLA-G+ EVT from term pregnancy compared to first trimester revealed their unique phenotypes, gene expression profiles, and differing capacities to increase regulatory T cells (Treg) during coculture assays, features that cannot be captured by using surrogate cell lines or animal models. Furthermore, clinical variables including gestational age and fetal sex significantly influenced EVT biology and function. These methods and approaches form a solid basis for further investigation of the role of HLA-G+ EVT in the development of detrimental placental inflammatory responses associated with pregnancy complications, including spontaneous preterm delivery and preeclampsia.
Collapse
|
20
|
Trophoblast cell influence on peripheral blood natural killer cell proliferation and phenotype in non-pregnant women and women in early pregnancy. Immunobiology 2020; 225:151910. [DOI: 10.1016/j.imbio.2020.151910] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/17/2020] [Accepted: 01/31/2020] [Indexed: 11/22/2022]
|
21
|
Tsunoda Y, Kudo M, Wada R, Ishino K, Kure S, Sakatani T, Takeshita T, Naito Z. Expression level of long noncoding RNA H19 of normotensive placentas in late pregnancy relates to the fetal growth restriction. J Obstet Gynaecol Res 2020; 46:1025-1034. [PMID: 32323427 DOI: 10.1111/jog.14260] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/02/2020] [Accepted: 04/04/2020] [Indexed: 11/28/2022]
Abstract
AIM Infants with fetal growth restriction (FGR) are at an increased risk of perinatal morbidity and mortality. The long noncoding RNA H19 gene is expressed abundantly in placental villi and recent studies suggest that it regulates FGR. However, the role of H19 in the FGR placenta remains unclear. This study aimed to clarify the relationship between H19 expression and FGR using normotensive placentas after 34 weeks of gestation. METHODS Formalin-fixed paraffin-embedded tissues from human placentas collected from pregnancies resulting in small for gestational age (SGA) and appropriate for gestational age (AGA) newborns were used. The histopathological features of placenta tissues, such as villous stromal fibrosis, the numbers of terminal villi, villous vessels and cytotrophoblasts were analyzed using hematoxylin and eosin, Masson's trichrome staining and immunostaining. The localization and expression of H19 in the placentas were demonstrated by in situ hybridization and reverse transcription-quantitative polymerase chain reaction (RT-qPCR), respectively. Moreover, the expression levels of H19-regulated molecules such as IGF2 and decorin (DCN) were measured by RT-qPCR. RESULTS Histopathological features of the placental villous were not different between placentas associated with SGA and AGA. H19 localized to the villous stroma, endothelial cells and cytotrophoblasts. Moreover, the expression level of H19 in SGA placentas was significantly lower than that in AGA placentas. The expression levels of IGF2 and DCN in SGA placentas tended to be lower than those in AGA placentas similarly to H19. CONCLUSION This study highlights the potential importance of regulatory events mediated by H19 in SGA placentas without histopathological abnormalities in late pregnancy.
Collapse
Affiliation(s)
- Youhei Tsunoda
- Department of Obstetrics and Gynecology, Nippon Medical School, Tokyo, Japan.,Department of Integrated Diagnostic Pathology, Nippon Medical School, Tokyo, Japan
| | - Mitsuhiro Kudo
- Department of Integrated Diagnostic Pathology, Nippon Medical School, Tokyo, Japan
| | - Ryuichi Wada
- Department of Integrated Diagnostic Pathology, Nippon Medical School, Tokyo, Japan
| | - Kousuke Ishino
- Department of Integrated Diagnostic Pathology, Nippon Medical School, Tokyo, Japan
| | - Shoko Kure
- Department of Integrated Diagnostic Pathology, Nippon Medical School, Tokyo, Japan
| | - Takashi Sakatani
- Department of Diagnostic Pathology, Nippon Medical School Hospital, Tokyo, Japan
| | - Toshiyuki Takeshita
- Department of Obstetrics and Gynecology, Nippon Medical School, Tokyo, Japan
| | - Zenya Naito
- Department of Integrated Diagnostic Pathology, Nippon Medical School, Tokyo, Japan
| |
Collapse
|
22
|
Begum G, O'Neill J, Chaudhary R, Blachford K, Snead DRJ, Berry M, Scott RAH, Logan A, Blanch RJ. Altered Decorin Biology in Proliferative Vitreoretinopathy: A Mechanistic and Cohort Study. Invest Ophthalmol Vis Sci 2019; 59:4929-4936. [PMID: 30347087 DOI: 10.1167/iovs.18-24299] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To determine if vitreous levels of the pro-fibrotic cytokine transforming growth factor beta2 (TGF-β2) and its opposing regulator decorin predict subsequent proliferative vitreoretinopathy (PVR) development in patients with rhegmatogenous retinal detachment (RRD). Methods We examined the effect of TGF-β2 and decorin on epithelial-mesenchymal transition (EMT) and collagen expression in vitro using ARPE-19 cells, and we analyzed extracellular matrix marker expression in PVR membrane and internal limiting membrane patient samples. We performed a prospective noninterventional cohort study, recruiting 125 patients undergoing vitrectomy for RRD and macular hole surgery, measured vitreous levels of TGF-β2 and decorin by ELISA, and followed them up for 6 months. Patients who did not develop PVR were compared to those who did, in order to determine whether vitreous TGF-β2 and decorin levels predicted PVR development. Results In vitro, TGF-β2 induced EMT and collagen production. Decorin strongly inhibited EMT and collagen production at high levels. PVR membranes expressed high levels of fibrosis-associated proteins, consistent with EMT. Vitreous TGF-β2 levels were unchanged between patients with macular holes and RRD who did or did not subsequently develop PVR. Average decorin levels were higher in the vitreous of RRD patients who subsequently developed PVR compared to those who did not, but at the measured vitreous concentrations (1-2 μg/mL), decorin did not demonstrate an in vitro inhibitory effect on EMT. Conclusions In vitro, high concentrations of decorin inhibited EMT and fibrosis. At the levels seen in human vitreous, decorin did not prevent fibrosis or EMT in vitro, and higher initial vitreous decorin levels were associated with the development of postoperative PVR after vitrectomy to treat RRD, but did not reliably predict the outcome.
Collapse
Affiliation(s)
- Ghazala Begum
- Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom.,NIHR Surgical Reconstruction and Microbiology Research Centre, University of Birmingham, Birmingham, United Kingdom
| | - Jenna O'Neill
- Ridgeway Research Ltd., St. Briavels, Gloucestershire, United Kingdom
| | - Rishika Chaudhary
- Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom.,NIHR Surgical Reconstruction and Microbiology Research Centre, University of Birmingham, Birmingham, United Kingdom.,Ophthalmology Department, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Karen Blachford
- Academic Unit of Ophthalmology, Birmingham and Midland Eye Centre, Sandwell and West Birmingham Hospitals NHS Trust, Birmingham, United Kingdom
| | - David R J Snead
- Department of Pathology, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, Warwickshire, United Kingdom
| | - Martin Berry
- Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Robert A H Scott
- Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom.,NIHR Surgical Reconstruction and Microbiology Research Centre, University of Birmingham, Birmingham, United Kingdom.,SpaMedica, Birmingham, United Kingdom
| | - Ann Logan
- Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom.,NIHR Surgical Reconstruction and Microbiology Research Centre, University of Birmingham, Birmingham, United Kingdom
| | - Richard J Blanch
- Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom.,NIHR Surgical Reconstruction and Microbiology Research Centre, University of Birmingham, Birmingham, United Kingdom.,Academic Unit of Ophthalmology, Birmingham and Midland Eye Centre, Sandwell and West Birmingham Hospitals NHS Trust, Birmingham, United Kingdom.,Academic Department of Military Surgery and Trauma, Royal Centre for Defence Medicine, Birmingham, United Kingdom
| |
Collapse
|
23
|
Pollheimer J, Vondra S, Baltayeva J, Beristain AG, Knöfler M. Regulation of Placental Extravillous Trophoblasts by the Maternal Uterine Environment. Front Immunol 2018; 9:2597. [PMID: 30483261 PMCID: PMC6243063 DOI: 10.3389/fimmu.2018.02597] [Citation(s) in RCA: 250] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 10/22/2018] [Indexed: 12/22/2022] Open
Abstract
During placentation invasive extravillous trophoblasts (EVTs) migrate into the maternal uterus and modify its vessels. In particular, remodeling of the spiral arteries by EVTs is critical for adapting blood flow and nutrient transport to the developing fetus. Failures in this process have been noticed in different pregnancy complications such as preeclampsia, intrauterine growth restriction, stillbirth, or recurrent abortion. Upon invasion into the decidua, the endometrium of pregnancy, EVTs encounter different maternal cell types such as decidual macrophages, uterine NK (uNK) cells and stromal cells expressing a plethora of growth factors and cytokines. Here, we will summarize development of the EVT lineage, a process occurring independently of the uterine environment, and formation of its different subtypes. Further, we will discuss interactions of EVTs with arteries, veins and lymphatics and illustrate how the decidua and its different immune cells regulate EVT differentiation, invasion and survival. The present literature suggests that the decidual environment and its soluble factors critically modulate EVT function and reproductive success.
Collapse
Affiliation(s)
- Jürgen Pollheimer
- Department of Obstetrics and Gynaecology, Medical University of Vienna, Vienna, Austria
| | - Sigrid Vondra
- Department of Obstetrics and Gynaecology, Medical University of Vienna, Vienna, Austria
| | - Jennet Baltayeva
- British Columbia's Children's Hospital Research Institute, Vancouver, BC, Canada.,Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, BC, Canada
| | - Alexander Guillermo Beristain
- British Columbia's Children's Hospital Research Institute, Vancouver, BC, Canada.,Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, BC, Canada
| | - Martin Knöfler
- Department of Obstetrics and Gynaecology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
24
|
Nandi P, Lim H, Torres-Garcia EJ, Lala PK. Human trophoblast stem cell self-renewal and differentiation: Role of decorin. Sci Rep 2018; 8:8977. [PMID: 29895842 PMCID: PMC5997742 DOI: 10.1038/s41598-018-27119-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 05/24/2018] [Indexed: 12/18/2022] Open
Abstract
The origin and regulation of stem cells sustaining trophoblast renewal in the human placenta remain unclear. Decorin, a leucine-rich proteoglycan restrains trophoblast proliferation, migration/invasiveness and endovascular differentiation, and local decorin overproduction is associated with preeclampsia (PE). Here, we tested the role of decorin in human trophoblast stem cell self-renewal and differentiation, using two models: an immortalized first trimester trophoblast cell line HTR-8/SVneo (HTR) and freshly isolated primary trophoblast (p-trophoblast) from early first trimester (6-9 weeks) placentas. Self-renewal capacity was measured by spheroid forming ability of single cells on ultra-low attachment plates for multiple generations. Markers of embryonic stem (ES) cells, trophoblast stem (TS) cells and trophoblast were used to identify stem cell hierarchy. Differentiation markers for syncytial and extravillous (EVT) pathways were employed to identify differentiated cells. Bewo cells were additionally used to explore DCN effects on syncytialization. Results reveal that the incidence of spheroid forming stem-like cells was 13-15% in HTR and 0.1-0.4%, in early first trimester p-trophoblast, including a stem cell hierarchy of two populations of ES and TS-like cells. DCN restrained ES cell self-renewal, promoted ES to TS transition and maintenance of TS cell stem-ness, but inhibited TS cell differentiation into both syncytial and EVT pathways.
Collapse
Affiliation(s)
- Pinki Nandi
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Hyobin Lim
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Eloy Jose Torres-Garcia
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Peeyush K Lala
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.
- Department of Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.
- Associate Scientist, Children's Health Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
25
|
Charkiewicz K, Jasinska E, Goscik J, Koc-Zorawska E, Zorawski M, Kuc P, Raba G, Kluz T, Kalinka J, Sakowicz A, Laudanski P. Angiogenic factor screening in women with mild preeclampsia - New and significant proteins in plasma. Cytokine 2017; 106:125-130. [PMID: 29111087 DOI: 10.1016/j.cyto.2017.10.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 10/19/2017] [Accepted: 10/23/2017] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The aim of this study was to analyse a panel of 60 angiogenic factors (pro-angiogenic and antiangiogenic) in the plasma of women with mild preeclampsia. MATERIALS AND METHODS We recruited 21 women between 25 and 40 weeks gestation with diagnosed mild preeclampsia into the study group and 27 healthy women with uncomplicated pregnancies of corresponding gestational age to that of the study to the control group. We used a quantitative protein macroarray method that allowed for analysis of 60 angiogenic proteins per sample simultaneously. RESULTS We showed a statistically significant increase in the concentration of 8 proteins, interferon gamma (IFN-γ), interleukin 6 (IL-6), leukaemia inhibitory factor (LIF), heparin-binding EGF-like growth factor (HB-EGF), hepatocyte growth factor (HGF), C-X-C motif chemokine 10 (IP-10), leptin and platelet-derived growth factor BB (PDGF-BB), as well as a significant decrease in the concentration of 3 proteins, vascular endothelial growth factor (VEGF), placental growth factor (PlGF) and follistatin, in the plasma of women with preeclampsia. CONCLUSION Based on our findings, it seems that protein factors may play an important role in the pathogenesis of preeclampsia, and there are many proteins that have not been studied in PE to date. There are no previous studies assessing the LIF, follistatin, HGF, HB-EGF and PDGF-BB concentrations in the plasma of women with PE; therefore, our obtained results indicate that these proteins are new factors that can play an important role in the pathomechanisms of PE.
Collapse
Affiliation(s)
- Karol Charkiewicz
- Department of Perinatology and Obstetrics, Medical University of Bialystok, Marii Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland.
| | - Elwira Jasinska
- Department of Perinatology and Obstetrics, Medical University of Bialystok, Marii Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Joanna Goscik
- Faculty of Computer Science, Bialystok University of Technology, Wiejska 45A, 15-351 Bialystok, Poland
| | - Ewa Koc-Zorawska
- Department of Nephrology and Hypertension with Dialysis Unit, Medical University of Bialystok, Poland, Marii Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Marcin Zorawski
- Department of Clinical Medicine, Medical University of Bialystok, Poland, Marii Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Pawel Kuc
- Department of Perinatology and Obstetrics, Medical University of Bialystok, Marii Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Grzegorz Raba
- Institute of Obstetric and Emergency Medicine, University of Rzeszow, Żurawica, 37-710 Podkarpackie, Poland
| | - Tomasz Kluz
- Department of Obstetrics and Gynecology, Fryderyk Chopin University Hospital No 1, Faculty of Medicine, Rzeszow University, Poland
| | - Jaroslaw Kalinka
- Department of Perinatology, Medical University of Lodz, 91-429 Lodz, Poland
| | - Agata Sakowicz
- Department of Medical Biotechnology, Medical University of Lodz, 91-425 Lodz, Poland
| | - Piotr Laudanski
- Department of Perinatology and Obstetrics, Medical University of Bialystok, Marii Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland.
| |
Collapse
|
26
|
Menkhorst E, Winship A, Van Sinderen M, Dimitriadis E. Human extravillous trophoblast invasion: intrinsic and extrinsic regulation. Reprod Fertil Dev 2017; 28:406-15. [PMID: 25163485 DOI: 10.1071/rd14208] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 07/27/2014] [Indexed: 12/12/2022] Open
Abstract
During the establishment of pregnancy, a human blastocyst implants into the uterine endometrium to facilitate the formation of a functional placenta. Implantation involves the blastocyst adhering to the uterine luminal epithelium before the primitive syncytiotrophoblast and subsequently specialised cells, the extravillous trophoblast (EVT), invade into the decidua in order to engraft and remodel uterine spiral arteries, creating the placental blood supply at the end of the first trimester. Defects in EVT invasion lead to abnormal placentation and thus adverse pregnancy outcomes. The local decidual environment is thought to play a key role in regulating trophoblast invasion. Here we describe the major cell types present in the decidua during the first trimester of pregnancy and review what is known about their regulation of EVT invasion. Overall, the evidence suggests that in a healthy pregnancy almost all cell types in the decidua actively promote EVT invasion and, further, that reduced EVT invasion towards the end of the first trimester is regulated, in part, by the reduced invasive capacity of EVTs shown at this time.
Collapse
Affiliation(s)
- E Menkhorst
- MIMR-PHI Institute of Medical Research, 27-31 Wright St, Clayton, Vic. 3168, Australia
| | - A Winship
- MIMR-PHI Institute of Medical Research, 27-31 Wright St, Clayton, Vic. 3168, Australia
| | - M Van Sinderen
- MIMR-PHI Institute of Medical Research, 27-31 Wright St, Clayton, Vic. 3168, Australia
| | - E Dimitriadis
- MIMR-PHI Institute of Medical Research, 27-31 Wright St, Clayton, Vic. 3168, Australia
| |
Collapse
|
27
|
Decorin over-expression by decidual cells in preeclampsia: a potential blood biomarker. Am J Obstet Gynecol 2016; 215:361.e1-361.e15. [PMID: 27001218 DOI: 10.1016/j.ajog.2016.03.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 03/10/2016] [Accepted: 03/11/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND Decorin, a leucine-rich proteoglycan that is produced by decidual cells, limits invasion and endovascular differentiation of extravillous trophoblast cells during early placentation by binding to multiple tyrosine kinase receptors, in particular, vascular endothelial growth factor receptor-2. OBJECTIVE Because many studies have reported an association between poor trophoblast invasion and endovascular differentiation with preeclampsia, the studies reported here tested (1) whether decorin over-expression in the chorionic villi and/or basal decidua is associated with preeclampsia and, if so, (2) whether this association results in a hypoinvasive placenta, and (3) whether elevated plasma decorin concentration in the second trimester is a predictive biomarker for preeclampsia. STUDY DESIGN Decorin messenger RNA expression was measured with quantitative polymerase chain reaction at the tissue level and with in situ hybridization at the cellular level using (35)S-labeled antisense complimentary RNA probe in placentas from healthy control subjects and subjects with preeclampsia (14 each, 23-40 weeks of gestation). Tissue sections of the same placentas were also immunostained for decorin protein. A decorin over-expressing human endometrial stromal cell line was tested for invasion-regulatory effects on an invasive first-trimester extravillous trophoblast cell line HTR-8/SVneo plated in cocultures that were separated by a semipermeable membrane. Furthermore, we conducted retrospective measurements of plasma decorin levels during the second trimester (15-18 weeks of gestation) in a cohort of 28 body mass index-matched pairs of control subjects and subjects with preeclampsia before the onset of clinical disease. RESULTS First, decorin messenger RNA expression at the cellular level measured with in situ hybridization exhibited profoundly higher expression levels in basal plate decidual cells within the placentas from preeclamptic subjects than those from control subjects at all gestational ages, whereas no difference between the 2 subject groups was noted in villus mesenchymal cells. Similarly decorin messenger RNA expression at the tissue level in chorionic villi (primarily resulting from fetally derived mesenchymal cells) did not differ significantly between control and preeclampsia placentas. These findings were validated with immunostaining for decorin protein. Second, knocking down decorin gene in a decorin over-expressing endometrial cell line (used as an in vitro surrogate of decorin over-expressing decidual cells) in cocultures with extravillous trophoblast cells abrogated its invasion-restraining actions on trophoblast cells, which indicated paracrine contribution of decorin over-expressing decidua to the poor trophoblast invasiveness in situ. Finally, retrospective measurement of plasma decorin levels during the second trimester in 28 body mass index-matched pairs of control subjects and subjects with preeclampsia revealed elevated plasma decorin levels in all subjects with preeclampsia in all body mass index groups. A receiver operating characteristic curve analysis revealed strong diagnostic performance of plasma decorin in the prediction of preeclampsia status. Although there was no significant gestational age-related change in decorin levels during the second trimester in control or subjects with preeclampsia, we found that plasma decorin had a significant inverse relationship with body mass index or bodyweight. CONCLUSION We conclude that decorin over-expression by basal decidual cells is associated with hypoinvasive phenotype and poor endovascular differentiation of trophoblast cells in preeclampsia and that elevated plasma decorin concentration is a potential predictive biomarker for preeclampsia before the onset of clinical signs.
Collapse
|
28
|
Decorin expression is decreased in first trimester placental tissue from pregnancies with small for gestation age infants at birth. Placenta 2016; 45:58-62. [DOI: 10.1016/j.placenta.2016.07.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 07/16/2016] [Accepted: 07/25/2016] [Indexed: 11/23/2022]
|
29
|
Cramer SF, Heller DS. Placenta Accreta and Placenta Increta: An Approach to Pathogenesis Based on the Trophoblastic Differentiation Pathway. Pediatr Dev Pathol 2016; 19:320-33. [PMID: 26492199 DOI: 10.2350/15-05-1641-oa.1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Morbid adherence remains a puzzling disease. This paper suggests that normal and morbidly adherent placentation may be viewed best in terms of trophoblastic stem cells and the mutually exclusive branches of the trophoblastic differentiation pathway-villous trophoblast (VT), interstitial and endovascular nonvillous trophoblast (NVT) at the implantation site, and a positional variation in the chorion. Based on cases of hysterectomies for morbid adherence seen over 30 years at a community hospital, analyzed with routine keratin stains, with actin and trichrome stains as indicated, and with attempts at ultrasonography-pathology correlation, we present selected observations. In true accreta, the site of morbid adherence was to dilated basal plate vessels infiltrated by endovascular NVT, with scant interstitial NVT, and normal myometrium. It appeared that excess blood flow into the placenta was due to excessively deep keratin-positive endovascular NVT that spread-independently of interstitial NVT-in an angiocentric fashion in both accreta and increta. Retroplacental abnormalities were due to myometrial destruction by interstitial NVT in increta, sometimes requiring actin stains for detection; and to an admixture of markedly dilated endometrial glands and vessels in true accreta, best appreciated with keratin stains. Variations of depth and extent in increta may be due to variations in myometrial tone, and in the protease-antiprotease balance. Morbidly adherent fetal membranes are described, and the role of caesarean section scars in incretas is addressed.
Collapse
Affiliation(s)
- Stewart F Cramer
- Department of Pathology, Rochester General Hospital, University of Rochester School of Medicine, Rochester, NY 14621, USA; and Department of Pathology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Debra S Heller
- Department of Pathology, Rochester General Hospital, University of Rochester School of Medicine, Rochester, NY 14621, USA; and Department of Pathology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| |
Collapse
|
30
|
Zhao L, Shao Q, Zhang Y, Zhang L, He Y, Wang L, Kong B, Qu X. Human monocytes undergo functional re-programming during differentiation to dendritic cell mediated by human extravillous trophoblasts. Sci Rep 2016; 6:20409. [PMID: 26857012 PMCID: PMC4746586 DOI: 10.1038/srep20409] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 01/06/2016] [Indexed: 01/02/2023] Open
Abstract
Maternal immune adaptation is required for a successful pregnancy to avoid rejection of the fetal–placental unit. Dendritic cells within the decidual microenvironment lock in a tolerogenic profile. However, how these tolerogenic DCs are induced and the underlying mechanisms are largely unknown. In this study, we show that human extravillous trophoblasts redirect the monocyte-to-DC transition and induce regulatory dendritic cells. DCs differentiated from blood monocytes in the presence of human extravillous trophoblast cell line HTR-8/SVneo displayed a DC-SIGN+CD14+CD1a− phenotype, similar with decidual DCs. HTR8-conditioned DCs were unable to develop a fully mature phenotype in response to LPS, and altered the cytokine secretory profile significantly. Functionally, conditioned DCs poorly induced the proliferation and activation of allogeneic T cells, whereas promoted CD4+CD25+Foxp3+ Treg cells generation. Furthermore, the supernatant from DC and HTR-8/SVneo coculture system contained significant high amount of M-CSF and MCP-1. Using neutralizing antibodies, we discussed the role of M-CSF and MCP-1 during monocyte-to-DCs differentiation mediated by extravillous trophoblasts. Our data indicate that human extravillous trophoblasts play an important role in modulating the monocyte-to-DC differentiation through M-CSF and MCP-1, which facilitate the establishment of a tolerogenic microenvironment at the maternal–fetal interface.
Collapse
Affiliation(s)
- Lei Zhao
- Institute of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, P.R. China
| | - Qianqian Shao
- Institute of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, P.R. China
| | - Yun Zhang
- Institute of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, P.R. China
| | - Lin Zhang
- Institute of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, P.R. China
| | - Ying He
- Institute of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, P.R. China
| | - Lijie Wang
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, P.R. China
| | - Beihua Kong
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, P.R. China
| | - Xun Qu
- Institute of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, 250012, Shandong, P.R. China
| |
Collapse
|
31
|
Xu J, Sivasubramaniyam T, Yinon Y, Tagliaferro A, Ray J, Nevo O, Post M, Caniggia I. Aberrant TGFβ Signaling Contributes to Altered Trophoblast Differentiation in Preeclampsia. Endocrinology 2016; 157:883-99. [PMID: 26653761 DOI: 10.1210/en.2015-1696] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
TGFβ has been implicated in preeclampsia, but its intracellular signaling via phosphorylated mothers against decapentaplegic (SMADs) and SMAD-independent proteins in the placenta remains elusive. Here we show that TGFβ receptor-regulated SMAD2 was activated (Ser(465/467) phosphorylation) in syncytiotrophoblast and proliferating extravillous trophoblast cells of first-trimester placenta, whereas inhibitory SMAD7 located primarily to cytotrophoblast cells. SMAD2 phosphorylation decreased with advancing gestation, whereas SMAD7 expression increased and shifted to syncytiotrophoblasts toward term. Additionally, we found that the TGFβ SMAD-independent signaling via partitioning defective protein 6 (PARD6)/Smad ubiquitylation regulatory factor was activated at approximately 10-12 weeks of gestation in cytotrophoblast and extravillous trophoblast cells comprising the anchoring column. Placentae from early-onset, but not late-onset, preeclampsia exhibited elevated SMAD2 phosphorylation and SMAD7 levels. Whereas PARD6 expression increased and SMURF1 levels decreased in preeclamptic placentae, their association increased. SMAD2 phosphorylation by TGFβ in villous explants and BeWo cells resulted in a reduction of Glial cell missing-1 (GCM1) and fusogenic protein syncytin-1 while increasing cell cycle regulators cyclin E-1 (CCNE1) and cyclin-dependent kinase 4. SMAD7 abrogated the proliferative effects of TGFβ. CCNE1 levels were increased in preeclamptic placentae, whereas GCM1 was markedly reduced. In addition, TGFβ treatment increased the association of PARD6 and SMURF1 and down-regulated Ras homolog gene family, member A (RHOA) GTPase in JEG3 cells. In a wound assay, TGFβ treatment increased the association of PARD6 and SMURF1 and triggered JEG3 cell migration through increased cellular protrusions. Taken together, our data indicate that TGFβ signaling via both SMAD2/7 and PARD6/SMURF1 pathways plays a role in trophoblast growth and differentiation. Altered SMAD regulation of GCM1 and CCNE1 and aberrant expression/activation of PARD6/SMURF1 may contribute to the pathogenesis of preeclampsia by affecting cellular pathways associated with this disorder.
Collapse
Affiliation(s)
- Jing Xu
- Lunenfeld-Tanenbaum Research Institute (J.X., T.S., A.T., J.R., I.C.), Mt Sinai Hospital, Toronto, Ontario, Canada M5T 3H7; Departments of Obstetrics and Gynecology (Y.Y., O.N., I.C.), Physiology (T.S., J.R., M.P., I.C.), and Pediatrics (M.P.), University of Toronto, Toronto, Ontario, Canada; and Institute of Medical Science (M.P.), The Hospital for Sick Children, Toronto, Ontario, Canada M5G 1X8
| | - Tharini Sivasubramaniyam
- Lunenfeld-Tanenbaum Research Institute (J.X., T.S., A.T., J.R., I.C.), Mt Sinai Hospital, Toronto, Ontario, Canada M5T 3H7; Departments of Obstetrics and Gynecology (Y.Y., O.N., I.C.), Physiology (T.S., J.R., M.P., I.C.), and Pediatrics (M.P.), University of Toronto, Toronto, Ontario, Canada; and Institute of Medical Science (M.P.), The Hospital for Sick Children, Toronto, Ontario, Canada M5G 1X8
| | - Yoav Yinon
- Lunenfeld-Tanenbaum Research Institute (J.X., T.S., A.T., J.R., I.C.), Mt Sinai Hospital, Toronto, Ontario, Canada M5T 3H7; Departments of Obstetrics and Gynecology (Y.Y., O.N., I.C.), Physiology (T.S., J.R., M.P., I.C.), and Pediatrics (M.P.), University of Toronto, Toronto, Ontario, Canada; and Institute of Medical Science (M.P.), The Hospital for Sick Children, Toronto, Ontario, Canada M5G 1X8
| | - Andrea Tagliaferro
- Lunenfeld-Tanenbaum Research Institute (J.X., T.S., A.T., J.R., I.C.), Mt Sinai Hospital, Toronto, Ontario, Canada M5T 3H7; Departments of Obstetrics and Gynecology (Y.Y., O.N., I.C.), Physiology (T.S., J.R., M.P., I.C.), and Pediatrics (M.P.), University of Toronto, Toronto, Ontario, Canada; and Institute of Medical Science (M.P.), The Hospital for Sick Children, Toronto, Ontario, Canada M5G 1X8
| | - Jocelyn Ray
- Lunenfeld-Tanenbaum Research Institute (J.X., T.S., A.T., J.R., I.C.), Mt Sinai Hospital, Toronto, Ontario, Canada M5T 3H7; Departments of Obstetrics and Gynecology (Y.Y., O.N., I.C.), Physiology (T.S., J.R., M.P., I.C.), and Pediatrics (M.P.), University of Toronto, Toronto, Ontario, Canada; and Institute of Medical Science (M.P.), The Hospital for Sick Children, Toronto, Ontario, Canada M5G 1X8
| | - Ori Nevo
- Lunenfeld-Tanenbaum Research Institute (J.X., T.S., A.T., J.R., I.C.), Mt Sinai Hospital, Toronto, Ontario, Canada M5T 3H7; Departments of Obstetrics and Gynecology (Y.Y., O.N., I.C.), Physiology (T.S., J.R., M.P., I.C.), and Pediatrics (M.P.), University of Toronto, Toronto, Ontario, Canada; and Institute of Medical Science (M.P.), The Hospital for Sick Children, Toronto, Ontario, Canada M5G 1X8
| | - Martin Post
- Lunenfeld-Tanenbaum Research Institute (J.X., T.S., A.T., J.R., I.C.), Mt Sinai Hospital, Toronto, Ontario, Canada M5T 3H7; Departments of Obstetrics and Gynecology (Y.Y., O.N., I.C.), Physiology (T.S., J.R., M.P., I.C.), and Pediatrics (M.P.), University of Toronto, Toronto, Ontario, Canada; and Institute of Medical Science (M.P.), The Hospital for Sick Children, Toronto, Ontario, Canada M5G 1X8
| | - Isabella Caniggia
- Lunenfeld-Tanenbaum Research Institute (J.X., T.S., A.T., J.R., I.C.), Mt Sinai Hospital, Toronto, Ontario, Canada M5T 3H7; Departments of Obstetrics and Gynecology (Y.Y., O.N., I.C.), Physiology (T.S., J.R., M.P., I.C.), and Pediatrics (M.P.), University of Toronto, Toronto, Ontario, Canada; and Institute of Medical Science (M.P.), The Hospital for Sick Children, Toronto, Ontario, Canada M5G 1X8
| |
Collapse
|
32
|
Lala PK, Nandi P. Mechanisms of trophoblast migration, endometrial angiogenesis in preeclampsia: The role of decorin. Cell Adh Migr 2016; 10:111-25. [PMID: 26745663 DOI: 10.1080/19336918.2015.1106669] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The objective of the present review is to synthesize the information on the cellular and molecular players responsible for maintaining a homeostatic balance between a naturally invasive human placenta and the maternal uterus in pregnancy; to review the roles of decorin (DCN) as a molecular player in this homeostasis; to list the common maladies associated with a break-down in this homeostasis, resulting from a hypo-invasive or hyper-invasive placenta, and their underlying mechanisms. We show that both the fetal components of the placenta, represented primarily by the extravillous trophoblast, and the maternal component represented primarily by the decidual tissue and the endometrial arterioles, participate actively in this balance. We discuss the process of uterine angiogenesis in the context of uterine arterial changes during normal pregnancy and preeclampsia. We compare and contrast trophoblast growth and invasion with the processes involved in tumorigenesis with special emphasis on the roles of DCN and raise important questions that remain to be addressed. Decorin (DCN) is a small leucine-rich proteoglycan produced by stromal cells, including dermal fibroblasts, chondrocytes, chorionic villus mesenchymal cells and decidual cells of the pregnant endometrium. It contains a 40 kDa protein core having 10 leucine-rich repeats covalently linked with a glycosaminoglycan chain. Biological functions of DCN include: collagen assembly, myogenesis, tissue repair and regulation of cell adhesion and migration by binding to ECM molecules or antagonising multiple tyrosine kinase receptors (TKR) including EGFR, IGF-IR, HGFR and VEGFR-2. DCN restrains angiogenesis by binding to thrombospondin-1, TGFβ, VEGFR-2 and possibly IGF-IR. DCN can halt tumor growth by antagonising oncogenic TKRs and restraining angiogenesis. DCN actions at the fetal-maternal interface include restraint of trophoblast migration, invasion and uterine angiogenesis. We demonstrate that DCN overexpression in the decidua is associated with preeclampsia (PE); this may have a causal role in PE by compromising endovascular differentiation of the trophoblast and uterine angiogenesis, resulting in poor arterial remodeling. Elevated DCN level in the maternal blood is suggested as a potential biomarker in PE.
Collapse
Affiliation(s)
- Peeyush K Lala
- a Department of Anatomy and Cell Biology , Schulich School of Medicine and Dentistry, the University of Western Ontario , London , Ontario , Canada.,b Department of Oncology , Schulich School of Medicine and Dentistry, the University of Western Ontario , London , Ontario , Canada.,c Chidren's Health Research Institute, Schulich School of Medicine and Dentistry, the University of Western Ontario , London , Ontario , Canada
| | - Pinki Nandi
- a Department of Anatomy and Cell Biology , Schulich School of Medicine and Dentistry, the University of Western Ontario , London , Ontario , Canada
| |
Collapse
|
33
|
Nandi P, Siddiqui MF, Lala PK. Restraint of Trophoblast Invasion of the Uterus by Decorin: Role in Pre-eclampsia. Am J Reprod Immunol 2015; 75:351-60. [DOI: 10.1111/aji.12449] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 10/16/2015] [Indexed: 11/28/2022] Open
Affiliation(s)
- Pinki Nandi
- Departments of Anatomy and Cell biology; Schulich School of Medicine and Dentistry; The University of Western Ontario; London ON Canada
| | - Mohammad Fyyaz Siddiqui
- Departments of Anatomy and Cell biology; Schulich School of Medicine and Dentistry; The University of Western Ontario; London ON Canada
| | - Peeyush K Lala
- Departments of Anatomy and Cell biology; Schulich School of Medicine and Dentistry; The University of Western Ontario; London ON Canada
- Department of Oncology; Schulich School of Medicine and Dentistry; The University of Western Ontario; London ON Canada
- Children's Health Research Institute; Schulich School of Medicine and Dentistry; The University of Western Ontario; London ON Canada
| |
Collapse
|
34
|
Winship A, Cuman C, Rainczuk K, Dimitriadis E. Fibulin-5 is upregulated in decidualized human endometrial stromal cells and promotes primary human extravillous trophoblast outgrowth. Placenta 2015; 36:1405-11. [PMID: 26506560 DOI: 10.1016/j.placenta.2015.10.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 09/25/2015] [Accepted: 10/12/2015] [Indexed: 01/01/2023]
Abstract
Interactions between the highly invasive trophoblasts and the maternal uterine decidual extracellular matrix (ECM) are crucial in the determination of a successful pregnancy. Fibulin-5 (FBLN5) is a member of the fibulin family that alters cell adhesive and invasive properties and is expressed in human villous cytotrophoblasts. We aimed to determine the expression and immunolocalization of FBLN5 in human first trimester decidua and examine the effect of FBLN5 in trophoblast invasion in vitro using a first trimester placental villous outgrowth assay. We demonstrated that FBLN5 mRNA expression is upregulated in response to cAMP-mediated decidualization of primary human endometrial stromal cells, although FBLN5 itself does not enhance decidualization. We reported for the first time, FBLN5 protein production in first trimester decidual cells and also co-localization to HLAG-positive EVTs in first trimester decidua. Consequently, we investigated the effects of exogenous FBLN5 on placental villous outgrowth in vitro and demonstrated that FBLN5 promotes EVT migration/invasion. This is the first study to identify FBLN5 in decidualized human endometrial stromal cells, first trimester decidua and EVT and determine a functional role for FBLN5 in human EVTs, suggesting that decidual and or EVT-derived FBLN5 regulates EVT invasion and placentation in women.
Collapse
Affiliation(s)
- Amy Winship
- Embryo Implantation Laboratory, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia
| | - Carly Cuman
- Embryo Implantation Laboratory, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Medicine, Monash University, Clayton, VIC, Australia
| | - Katarzyna Rainczuk
- Embryo Implantation Laboratory, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Evdokia Dimitriadis
- Embryo Implantation Laboratory, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC, Australia; Department of Molecular and Translational Medicine, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
35
|
Abstract
The hydatidiform mole (HM) is a placental pathology of androgenetic origin. Placental villi have an abnormal hyperproliferation event and hydropic degeneration. Three situations can be envisaged at its origin: 1. The destruction/expulsion of the female pronucleus at the time of fertilization by 1 or 2 spermatozoa with the former being followed by an endoreplication of the male pronucleus leading to a complete hydatidiform mole (CHM) 2. A triploid zygote (fertilization by 2 spermatozoa) leading to a partial hydatidiform mole (PHM) but can also lead to haploid and diploid clones. The diploid clone may produce a normal fetus while the haploid clone after endoreplication generates a CHM 3. A nutritional defect during the differentiation of the oocytes or the deterioration of the limited oxygen pressure during the first trimester of gestation may lead to the formation of a HM. In countries with poor medical health care system, moles (mainly the CHM) can become invasive or, in rare cases, lead to gestational choriocarcinomas.
Collapse
Affiliation(s)
- Jean-Jacques Candelier
- a Unité 1197 INSERM, Stem cell-niches Interactions: Physiology , Tumors and Tissular Repair, Hôpital Paul Brousse, Bâtiment Lavoisier , Villejuif , France.,b University of Paris-Saclay , Saint-Aubin , France
| |
Collapse
|
36
|
Chen CP. Placental villous mesenchymal cells trigger trophoblast invasion. Cell Adh Migr 2015; 8:94-7. [PMID: 24622731 DOI: 10.4161/cam.28347] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The successful transformation of uterine spiral arteries by invasion trophoblasts is critical for the formation of the human hemochorial placenta. Placental trophoblast migration and invasion are well regulated by various autocrine/paracrine factors at maternal-fetal interface. Human placental multipotent mesenchymal stromal cells (hPMSCs) are a subpopulation of villous mesenchymal cells and have been shown to produce a wide array of soluble cytokines and growth factors including HGF (hepatocyte growth factor). The function of hPMSCs in placental villous microenvironment has not been explored. The interaction between hPMSCs and trophoblasts was proposed in vitro in a recent article. HGF produced by hPMSCs was able to engage c-Met receptor on trophoblast and induced the trophoblast cAMP expression. The cAMP activated PKA, which in turn, signaled to Rap1 and led to integrin ?1 activation. The total integrin ?1 protein expression by trophoblasts was not affected by HGF stimulation. Hypoxia downregulated HGF expression by hPMSCs. HGF and PKA activator 6-Bnz-cAMP increased trophoblast adhesion and migration that were inhibited by PKA inhibitor H89 or Rap1 siRNA. Thus, hPMSCs-derived paracrine HGF can regulate trophoblast migration during placentation. These findings provided insight revealing at least one mechanism by which hPMSCs implicated in the development of preeclampsia.
Collapse
Affiliation(s)
- Chie-Pein Chen
- Division of High Risk Pregnancy; Mackay Memorial Hospital; Taipei, Taiwan; Department of Medical Research; Mackay Memorial Hospital; Taipei, Taiwan
| |
Collapse
|
37
|
Human HLA-G+ extravillous trophoblasts: Immune-activating cells that interact with decidual leukocytes. Proc Natl Acad Sci U S A 2015; 112:7219-24. [PMID: 26015573 DOI: 10.1073/pnas.1507977112] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Invading human leukocyte antigen-G+ (HLA-G+) extravillous trophoblasts (EVT) are rare cells that are believed to play a key role in the prevention of a maternal immune attack on foreign fetal tissues. Here highly purified HLA-G+ EVT and HLA-G- villous trophoblasts (VT) were isolated. Culture on fibronectin that EVT encounter on invading the uterus increased HLA-G, EGF-Receptor-2, and LIF-Receptor expression on EVT, presumably representing a further differentiation state. Microarray and functional gene set enrichment analysis revealed a striking immune-activating potential for EVT that was absent in VT. Cocultures of HLA-G+ EVT with sample matched decidual natural killer cells (dNK), macrophages, and CD4+ and CD8+ T cells were established. Interaction of EVT with CD4+ T cells resulted in increased numbers of CD4+CD25(HI)FOXP3+CD45RA+ resting regulatory T cells (Treg) and increased the expression level of the Treg-specific transcription factor FOXP3 in these cells. However, EVT did not enhance cytokine secretion in dNK, whereas stimulation of dNK with mitogens or classical natural killer targets confirmed the distinct cytokine secretion profiles of dNK and peripheral blood NK cells (pNK). EVT are specialized cells involved in maternal-fetal tolerance, the properties of which are not imitated by HLA-G-expressing surrogate cell lines.
Collapse
|
38
|
Watkins AJ, Lucas ES, Marfy-Smith S, Bates N, Kimber SJ, Fleming TP. Maternal nutrition modifies trophoblast giant cell phenotype and fetal growth in mice. Reproduction 2015; 149:563-75. [PMID: 25755287 PMCID: PMC4418750 DOI: 10.1530/rep-14-0667] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 03/09/2015] [Indexed: 01/15/2023]
Abstract
Mammalian placentation is dependent upon the action of trophoblast cells at the time of implantation. Appropriate fetal growth, regulated by maternal nutrition and nutrient transport across the placenta, is a critical factor for adult offspring long-term health. We have demonstrated that a mouse maternal low-protein diet (LPD) fed exclusively during preimplantation development (Emb-LPD) increases offspring growth but programmes adult cardiovascular and metabolic disease. In this study, we investigate the impact of maternal nutrition on post-implantation trophoblast phenotype and fetal growth. Ectoplacental cone explants were isolated at day 8 of gestation from female mice fed either normal protein diet (NPD: 18% casein), LPD (9% casein) or Emb-LPD and cultured in vitro. We observed enhanced spreading and cell division within proliferative and secondary trophoblast giant cells (TGCs) emerging from explants isolated from LPD-fed females when compared with NPD and Emb-LPD explants after 24 and 48 h. Moreover, both LPD and Emb-LPD explants showed substantial expansion of TGC area during 24–48 h, not observed in NPD. No difference in invasive capacity was observed between treatments using Matrigel transwell migration assays. At day 17 of gestation, LPD- and Emb-LPD-fed conceptuses displayed smaller placentas and larger fetuses respectively, resulting in increased fetal:placental ratios in both groups compared with NPD conceptuses. Analysis of placental and yolk sac nutrient signalling within the mammalian target of rapamycin complex 1 pathway revealed similar levels of total and phosphorylated downstream targets across groups. These data demonstrate that early post-implantation embryos modify trophoblast phenotype to regulate fetal growth under conditions of poor maternal nutrition.
Collapse
Affiliation(s)
- Adam J Watkins
- Centre for Biological SciencesSouthampton General Hospital, University of Southampton, Southampton SO16 6YD, UKSchool of Life and Health SciencesAston Research Centre for Healthy Ageing, Aston University, Birmingham B4 7ET, UKFaculty of Life SciencesUniversity of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK Centre for Biological SciencesSouthampton General Hospital, University of Southampton, Southampton SO16 6YD, UKSchool of Life and Health SciencesAston Research Centre for Healthy Ageing, Aston University, Birmingham B4 7ET, UKFaculty of Life SciencesUniversity of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| | - Emma S Lucas
- Centre for Biological SciencesSouthampton General Hospital, University of Southampton, Southampton SO16 6YD, UKSchool of Life and Health SciencesAston Research Centre for Healthy Ageing, Aston University, Birmingham B4 7ET, UKFaculty of Life SciencesUniversity of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| | - Stephanie Marfy-Smith
- Centre for Biological SciencesSouthampton General Hospital, University of Southampton, Southampton SO16 6YD, UKSchool of Life and Health SciencesAston Research Centre for Healthy Ageing, Aston University, Birmingham B4 7ET, UKFaculty of Life SciencesUniversity of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| | - Nicola Bates
- Centre for Biological SciencesSouthampton General Hospital, University of Southampton, Southampton SO16 6YD, UKSchool of Life and Health SciencesAston Research Centre for Healthy Ageing, Aston University, Birmingham B4 7ET, UKFaculty of Life SciencesUniversity of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| | - Susan J Kimber
- Centre for Biological SciencesSouthampton General Hospital, University of Southampton, Southampton SO16 6YD, UKSchool of Life and Health SciencesAston Research Centre for Healthy Ageing, Aston University, Birmingham B4 7ET, UKFaculty of Life SciencesUniversity of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| | - Tom P Fleming
- Centre for Biological SciencesSouthampton General Hospital, University of Southampton, Southampton SO16 6YD, UKSchool of Life and Health SciencesAston Research Centre for Healthy Ageing, Aston University, Birmingham B4 7ET, UKFaculty of Life SciencesUniversity of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| |
Collapse
|
39
|
Guillomot M, Campion E, Prézelin A, Sandra O, Hue I, Le Bourhis D, Richard C, Biase F, Rabel C, Wallace R, Lewin H, Renard JP, Jammes H. Spatial and temporal changes of Decorin, Type I collagen and Fibronectin expression in normal and clone bovine placenta. Placenta 2014; 35:737-47. [DOI: 10.1016/j.placenta.2014.06.366] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 06/12/2014] [Accepted: 06/13/2014] [Indexed: 01/06/2023]
|
40
|
Lockwood CJ, Basar M, Kayisli UA, Guzeloglu-Kayisli O, Murk W, Wang J, De Paz N, Shapiro JP, Masch RJ, Semerci N, Huang SJ, Schatz F. Interferon-γ protects first-trimester decidual cells against aberrant matrix metalloproteinases 1, 3, and 9 expression in preeclampsia. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:2549-59. [PMID: 25065683 DOI: 10.1016/j.ajpath.2014.05.025] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 04/23/2014] [Accepted: 05/28/2014] [Indexed: 01/19/2023]
Abstract
Human extravillous trophoblast (EVT) invades the decidua via integrin receptors and subsequently degrades extracellular matrix proteins. In preeclampsia (PE), shallow EVT invasion elicits incomplete spiral artery remodeling, causing reduced uteroplacental blood flow. Previous studies show that preeclamptic decidual cells, but not interstitial EVTs, display higher levels of extracellular matrix-degrading matrix metalloproteinase (MMP)-9, but not MMP-2. Herein, we extend our previous PE-related assessment of MMP-2 and MMP-9 to include MMP-1, which preferentially degrades fibrillar collagens, and MMP-3, which can initiate a local proteolytic cascade. In human first-trimester decidual cells incubated with estradiol, tumor necrosis factor-α (TNF-α) significantly enhanced MMP-1, MMP-3, and MMP-9 mRNA and protein levels and activity measured by real-time quantitative RT-PCR, ELISA, immunoblotting, and zymography, respectively. In contrast, interferon γ (IFN-γ) reversed these effects and medroxyprogesterone acetate elicited further reversal. Immunoblotting revealed that p38 mitogen-activated protein kinase signaling mediated TNF-α enhancement of MMP-1, MMP-3, and MMP-9, whereas IFN-γ inhibited p38 mitogen-activated protein kinase phosphorylation. Unlike highly regulated MMP-1, MMP-3, and MMP-9, MMP-2 mRNA and protein expression was constitutive in decidual cells. Because inflammation underlies PE-associated shallow EVT invasion, these results suggest that excess macrophage-derived TNF-α augments expression of MMP-1, MMP-3, and MMP-9 in decidual cells to interfere with normal stepwise EVT invasion of the decidua. In contrast, decidual natural killer cell-derived IFN-γ reverses such TNF-α-induced MMPs to protect against PE.
Collapse
Affiliation(s)
- Charles J Lockwood
- Department of Obstetrics and Gynecology, The Ohio State University College of Medicine, Columbus, Ohio.
| | - Murat Basar
- Department of Obstetrics and Gynecology, The Ohio State University College of Medicine, Columbus, Ohio
| | - Umit A Kayisli
- Department of Obstetrics and Gynecology, The Ohio State University College of Medicine, Columbus, Ohio
| | - Ozlem Guzeloglu-Kayisli
- Department of Obstetrics and Gynecology, The Ohio State University College of Medicine, Columbus, Ohio
| | - William Murk
- Department of Chronic Disease Epidemiology, School of Public Health, Yale University, New Haven, Connecticut
| | - Jenny Wang
- Department of Obstetrics and Gynecology, School of Medicine, Yale University, New Haven, Connecticut
| | - Nicole De Paz
- Department of Obstetrics and Gynecology, School of Medicine, Yale University, New Haven, Connecticut
| | - John P Shapiro
- Department of Obstetrics and Gynecology, The Ohio State University College of Medicine, Columbus, Ohio
| | - Rachel J Masch
- Beth Israel Medical Center, Albert Einstein College of Medicine, New York, New York
| | - Nihan Semerci
- Department of Obstetrics and Gynecology, The Ohio State University College of Medicine, Columbus, Ohio
| | - S Joseph Huang
- Department of Obstetrics and Gynecology, The Ohio State University College of Medicine, Columbus, Ohio
| | - Frederick Schatz
- Department of Obstetrics and Gynecology, The Ohio State University College of Medicine, Columbus, Ohio
| |
Collapse
|
41
|
Chui A, Murthi P, Gunatillake T, Brennecke SP, Ignjatovic V, Monagle PT, Whitelock JM, Said JM. Altered decorin leads to disrupted endothelial cell function: a possible mechanism in the pathogenesis of fetal growth restriction? Placenta 2014; 35:596-605. [PMID: 24947404 DOI: 10.1016/j.placenta.2014.05.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 05/14/2014] [Accepted: 05/24/2014] [Indexed: 01/18/2023]
Abstract
OBJECTIVE Fetal growth restriction (FGR) is a key cause of adverse pregnancy outcome where maternal and fetal factors are identified as contributing to this condition. Idiopathic FGR is associated with altered vascular endothelial cell functions. Decorin (DCN) has important roles in the regulation of endothelial cell functions in vascular environments. DCN expression is reduced in FGR. The objectives were to determine the functional consequences of reduced DCN in a human microvascular endothelial cell line model (HMVEC), and to determine downstream targets of DCN and their expression in primary placental microvascular endothelial cells (PLECs) from control and FGR-affected placentae. APPROACH Short-interference RNA was used to reduce DCN expression in HMVECs and the effect on proliferation, angiogenesis and thrombin generation was determined. A Growth Factor PCR Array was used to identify downstream targets of DCN. The expression of target genes in control and FGR PLECs was performed. RESULTS DCN reduction decreased proliferation and angiogenesis but increased thrombin generation with no effect on apoptosis. The array identified three targets of DCN: FGF17, IL18 and MSTN. Validation of target genes confirmed decreased expression of VEGFA, MMP9, EGFR1, IGFR1 and PLGF in HMVECs and PLECs from control and FGR pregnancies. CONCLUSIONS Reduction of DCN in vascular endothelial cells leads to disrupted cell functions. The targets of DCN include genes that play important roles in angiogenesis and cellular growth. Therefore, differential expression of these may contribute to the pathogenesis of FGR and disease states in other microvascular circulations.
Collapse
Affiliation(s)
- A Chui
- NorthWest Academic Centre, The University of Melbourne and Sunshine Hospital, PO Box 294, 176 Furlong Road, St Albans 3021, Australia.
| | - P Murthi
- Department of Perinatal Medicine, Pregnancy Research Centre, The Royal Women's Hospital, The University of Melbourne, Parkville 3052, Australia; Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville 3052, Australia
| | - T Gunatillake
- NorthWest Academic Centre, The University of Melbourne and Sunshine Hospital, PO Box 294, 176 Furlong Road, St Albans 3021, Australia; Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville 3052, Australia
| | - S P Brennecke
- Department of Perinatal Medicine, Pregnancy Research Centre, The Royal Women's Hospital, The University of Melbourne, Parkville 3052, Australia; Department of Obstetrics and Gynaecology, The University of Melbourne, Parkville 3052, Australia
| | - V Ignjatovic
- Murdoch Children's Research Institute, The Royal Children's Hospital and The University of Melbourne, Parkville 3052, Australia; Department of Clinical Haematology, The Royal Children's Hospital and The University of Melbourne, Parkville 3052, Australia; Department of Paediatrics, The Royal Children's Hospital and The University of Melbourne, Parkville 3052, Australia
| | - P T Monagle
- Murdoch Children's Research Institute, The Royal Children's Hospital and The University of Melbourne, Parkville 3052, Australia; Department of Clinical Haematology, The Royal Children's Hospital and The University of Melbourne, Parkville 3052, Australia; Department of Paediatrics, The Royal Children's Hospital and The University of Melbourne, Parkville 3052, Australia
| | - J M Whitelock
- Graduate School of Biomedical Engineering, University of New South Wales, Kensington 2033, Australia
| | - J M Said
- NorthWest Academic Centre, The University of Melbourne and Sunshine Hospital, PO Box 294, 176 Furlong Road, St Albans 3021, Australia
| |
Collapse
|
42
|
Cağlar M, Yavuzcan A, Göksu M, Alkan Bulbul G, Sıtkı Isenlik B, Ustün Y, Aydin S, Kumru S. Decorin: a possible marker for fetal growth restriction. Gynecol Endocrinol 2014; 30:141-4. [PMID: 24256371 DOI: 10.3109/09513590.2013.860125] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The aim of this study was to compare decorin (DCN) levels between pregnancies complicated by idiopathic fetal growth restriction (FGR) and uncomplicated pregnancies and to determine the relationship between DCN levels and clinical parameters. The study population consisted of two groups: control group consisted of 13 women with uncomplicated singleton pregnancies in the third trimester. Study group consisted of 14 singleton pregnancies complicated by idiopathic FGR who were admitted to the hospital for delivery in the third trimester of pregnancy. Maternal and fetal DCN levels were measured. Color Doppler flow assessments were performed. Relationship between DCN levels and clinical parameters was determined. Maternal DCN serum levels were significantly higher in complicated pregnancies by idiopathic FGR (p = 0.01). A statistically significant negative correlation was observed between maternal DCN serum levels and neonatal birth weight (r = -0.0506; p = 0.007). There was a significant correlation between umbilical artery (UA) DCN levels and UA S/D ratio (r = 0.512; p = 0.006) and UA RI (r = 0.405; p = 0.036). The risk of high DCN maternal serum levels (>7986.6 pg/mL) in pregnancy complicated by FGR was 8.25 times higher (RR = 8.25; 95% CI, 1.4-46.8). The results of our study showed that the presence of increased DCN levels in women with FGR could contribute to pathogenesis of the disease.
Collapse
Affiliation(s)
- Mete Cağlar
- Department of Obstetrics and Gynecology, Faculty of Medicine, Düzce University, Düzce , Turkey
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Borbely AU, Daher S, Ishigai MM, Mattar R, Sun SY, Knöfler M, Bevilacqua E, Oliveira SF. Decorin and biglycan immunolocalization in non-villous structures of healthy and pathological human placentas. Histopathology 2014; 64:616-25. [PMID: 24117774 DOI: 10.1111/his.12304] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 09/30/2013] [Indexed: 12/26/2022]
Abstract
AIMS Decorin and biglycan are members of the small leucine-rich proteoglycan family, and constituents of both the extracellular matrix (ECM) and the cell surface. They are recognized as important factors in the control of proliferation, migration and invasion in vivo and in vitro. In this study, the localization patterns of decorin and biglycan were determined in healthy placentas and in highly invasive placental pathologies. METHODS AND RESULTS The study included immunolocalization of decorin and biglycan in samples of first-trimester and term placentas, placenta accreta, invasive mole, and choriocarcinoma. Extravillous cytotrophoblast (EVT) cells were positive for both proteoglycans in all pathologies and in first-trimester placentas, although not in term placentas. Biglycan was immunolocalized in the ECM of all healthy and pathological placentas, whereas decorin was observed only in term placenta ECM. CONCLUSIONS The expression of both proteoglycans was cell-specific and gestation time-dependent in healthy placentas, and was associated with invasive EVT cells in pathological placentas. In view of the biological properties of these molecules, it is possible that the biglycan pattern found here is intrinsically implicated in the invasive activity of EVT cells in both healthy and disordered placentas.
Collapse
Affiliation(s)
- Alexandre U Borbely
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Lodhi RSZ, Nakabayashi K, Suzuki K, Yamada AY, Hazama R, Ebina Y, Yamada H. Relaxin has anti-apoptotic effects on human trophoblast-derived HTR-8/SV neo cells. Gynecol Endocrinol 2013; 29:1051-4. [PMID: 24070111 DOI: 10.3109/09513590.2013.829444] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The study was conducted to evaluate the effects of human relaxin on apoptosis in the human trophoblast derived HTR-8/SV neo cell line, which is a possible model of human extravillous trophoblasts (EVTs). HTR-8/SV neo cells, cultured in phenol red free RPMI1640 medium, were treated with different doses of human recombinant (rH2) relaxin in serum-deprived conditions. RT-PCR was used for evaluating relaxin receptor: RXFP1 and RXFP2 expression in HTR-8/SV neo cells. The cell death was examined by TUNEL assay. Furthermore, we investigated caspase-3, cleaved PARP and Bcl-2 expressions by Western blot analysis to recognize the translational effects of anti-apoptotic and pro-apoptotic proteins. RXFP1 and RXFP2 mRNA expression was observed in HTR-8/SV neo cells. Compared with untreated control cultures, treatment with rH2 relaxin, decreased TUNEL-positive rate in HTR-8/SV neo cells was observed. Western blot analysis revealed that treatment with rH2 relaxin decreased the expression of caspase-3 and cleaved PARP, but in contrast increased Bcl-2 expression in those cells. These results suggest that rH2 relaxin has anti-apoptotic effects on HTR8/SV neo cells by decreasing pro-apoptotic caspase-3 and cleaved PARP expression and up-regulating anti-apoptotic Bcl-2 expression.
Collapse
Affiliation(s)
- Romana S Z Lodhi
- Department of Obstetrics and Gynecology, Kobe University Graduate School of Medicine , Kobe , Japan
| | | | | | | | | | | | | |
Collapse
|
45
|
Van Sinderen M, Cuman C, Winship A, Menkhorst E, Dimitriadis E. The chrondroitin sulfate proteoglycan (CSPG4) regulates human trophoblast function. Placenta 2013; 34:907-12. [PMID: 23953863 DOI: 10.1016/j.placenta.2013.07.065] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 07/19/2013] [Accepted: 07/23/2013] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Trophoblast growth and invasion of the uterine endometrium are critical events during placentation and are tightly regulated by locally produced factors. Abnormal placentation can result in early miscarriage or preeclampsia and intrauterine growth restriction, leading to impaired fetal and/or maternal health. Chondroitin sulfate proteoglycan 4 (CSPG4) is involved in cancer cell migration and invasion, processes which are critical during placentation but unlike in cancer, trophoblast invasion is highly regulated. CSPG4 expression and function in trophoblast is unknown. We determined CSPG4 expression in human first trimester placenta and implantation sites, and investigated whether CSPG4 influenced proliferation, migration and invasion of a human extravillous trophoblast (EVT) cell line (HTR8/SVneo cells) as a model for extravillous trophoblast (EVT). METHODS AND RESULTS Immunoreactive CSPG4 localized to EVT cells in the trophoblast shell, subpopulations of interstitial EVT cells within the decidua and cytotrophoblast cells in placental villi. In HTR8/SVneo cells, siRNA knockdown of CSPG4 stimulated proliferation and decreased migration/invasion. In primary first trimester placental villi explants two cytokines, interleukin 11 (IL11) and leukemia inhibitory factor (LIF) with known roles in trophoblast function, stimulated CSPG4 mRNA expression and immunoreactive protein in the cyotrophoblast. DISCUSSION AND CONCLUSION This is the first demonstration of the production and function of CSPG4 in human placentation. These data suggest that locally produced CSPG4 stimulates human EVT migration and invasion and suggests that IL11 and LIF regulate villous cytotrophoblast differentiation towards the invasive phenotype at least in part via CSPG4.
Collapse
Affiliation(s)
- M Van Sinderen
- Prince Henry's Institute of Medical Research, Clayton, Victoria 3168, Australia
| | | | | | | | | |
Collapse
|
46
|
Warner JA, Zwezdaryk KJ, Day B, Sullivan DE, Pridjian G, Morris CA. Human cytomegalovirus infection inhibits CXCL12- mediated migration and invasion of human extravillous cytotrophoblasts. Virol J 2012; 9:255. [PMID: 23116176 PMCID: PMC3545970 DOI: 10.1186/1743-422x-9-255] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 10/29/2012] [Indexed: 01/18/2023] Open
Abstract
Background During the first trimester of pregnancy, a series of tightly regulated interactions govern the formation of a highly invasive population of fetal-derived extravillous cytotrophoblasts (EVT). Successful pregnancy is dependent on efficient invasion of the uterine wall and maternal spiral arteries by EVT. Dysregulated trophoblast invasion is associated with intrauterine growth restriction, birth defects, spontaneous abortion and preeclampsia. A number of soluble growth factors, cytokines, and chemokines modulate this process, fine-tuning the temporal and spatial aspects of cytotrophoblast invasion. In particular, the CXCL12/CXCR4 axis has been shown to specifically modulate cytotrophoblast differentiation, invasion, and survival throughout early pregnancy. Infection with human cytomegalovirus (HCMV) has been associated with impaired differentiation of cytotrophoblasts down the invasive pathway, specifically dysregulating the response to mitogens including epidermal growth factor (EGF) and hepatocyte growth factor (HGF). In this study, the effect of HCMV infection on the CXCL12-mediated migration and invasion of the EVT cell line SGHPL-4 was investigated. Results Infection with HCMV significantly decreased secretion of CXCL12 by SGHPL-4 cells, and induced a striking perinuclear accumulation of the chemokine. HCMV infection significantly increased mRNA and total cell surface expression of the two known receptors for CXCL12: CXCR4 and CXCR7. Functionally, HCMV-infected SGHPL-4 cells were unable to migrate or invade in response to a gradient of soluble CXCL12 in transwell assays. Conclusions Collectively, these studies demonstrate that HCMV impairs EVT migration and invasion induced by CXCL12. As HCMV has the ability to inhibit EVT migration and invasion through dysregulation of other relevant signaling pathways, it is likely that the virus affects multiple signaling pathways to impair placentation and contribute to some of the placental defects seen in HCMV-positive pregnancies.
Collapse
Affiliation(s)
- Jessica A Warner
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | | | | | | | | | | |
Collapse
|
47
|
Lala N, Girish GV, Cloutier-Bosworth A, Lala PK. Mechanisms in decorin regulation of vascular endothelial growth factor-induced human trophoblast migration and acquisition of endothelial phenotype. Biol Reprod 2012; 87:59. [PMID: 22699486 DOI: 10.1095/biolreprod.111.097881] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Extravillous trophoblast (EVT) cells of the human placenta invade the uterine decidua and utero-placental arteries to establish an efficient exchange of key molecules between maternal and fetal blood. Trophoblast invasion is stringently regulated in situ both positively and negatively by a variety of factors at the fetal-maternal interface to maintain a healthy utero-placental homeostasis. One such factor, decorin, a transforming growth factor (TGF)-beta binding, leucine-rich proteoglycan produced by the decidua, negatively regulates EVT proliferation, migration, and invasiveness independent of TGF-beta. We reported that these decorin actions were mediated by its binding to multiple tyrosine kinase receptors, including vascular endothelial growth factor receptor (VEGFR)-2. The present study explores the mechanisms underlying decorin antagonism of VEGF (VEGF-A) stimulation of endovascular differentiation of EVT using our EVT cell line, HTR-8/SVneo. We observe that decorin inhibits VEGF-induced EVT cell migration and endothelial-like tube formation on matrigel. VEGF activates MAPKs (p38 MAPK, MEK3/6, and ERK1/2) in EVT cells, and the activation is blocked in both cases by decorin. Employing selective MAPK inhibitors, we show that both p38 and ERK pathways contribute independently to VEGF-induced EVT migration and capillary-like tube formation. VEGF upregulates the vascular endothelial (VE) markers VE-cadherin and beta-catenin in EVT and endothelial cells, and this upregulation is blocked by decorin and MAPK inhibitors. These results suggest that decorin inhibits VEGF-A stimulation of trophoblast migration and endovascular differentiation by interfering with p38 MAPK and ERK1/2 activation. Thus decorin-mediated dual impediment of endovascular differentiation of the EVT and angiogenesis may have implications for pathogenesis of preeclampsia, a hypoinvasive trophoblast disorder in pregnancy.
Collapse
Affiliation(s)
- Neena Lala
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | | | | | | |
Collapse
|
48
|
Zong L, Gou W, Shao W, Huang P, Li C. Changes in the level of serum high-temperature requirement A1 (HtrA1) during pregnancy and its relationship to preeclampsia. Hypertens Pregnancy 2012; 31:389-97. [PMID: 22582805 DOI: 10.3109/10641955.2012.667472] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To investigate the expression and changes of high-temperature requirement A1 (HtrA1) during pregnancy and the use of this value in predicting preeclampsia. METHOD Serum samples were collected from pregnant mothers at different gestational weeks, and double-antibody sandwich enzyme-linked immunosorbent assay was employed to describe the changes in HtrA1 in serum during pregnancy. RESULTS (i) In Xi'an area of China, the incidence of preeclampsia was 4.95%, including 0.85% of early-onset type and 4.10% of late-onset type; (ii) the HtrA1 showed a lognormal distribution during pregnancy in the maternal serum, with the peak at 17-20 weeks of pregnancy; (iii) the HtrA1 levels in preeclampsia mothers peaked at 13-16 weeks of pregnancy, followed by acute decline until 21-24 weeks, then remained stable; (iv) the HtrA1 levels in preeclampsia mothers were higher than the control group in 13-16 weeks and lower than that in 21-24 weeks (p < 0.05); (v) the criteria using lgHtrA1 level at 1.684 during 13-16 weeks of pregnancy could predict hypertension disorders complicating pregnancy (HDCP) with sensitivity of 62.1% and specificity of 53.7%. This could be improved to 85 and 83%, respectively, when combined with body mass index as well as education background of the mother. CONCLUSION (i) HtrA1 showed lognormal distribution during pregnancy for all populations; (ii) HtrA1 level peaked at 17-20 weeks in normal group of pregnant mothers, and the increase of HtrA1 level in 13-16 weeks could predict the risk of preeclampsia; (iii) the risk calculation formula for preeclampsia: p (%) = eY/(1 + eY) (Y = -15.87 + 3.706 × lgHtrA1 + 0.134 × mean arterial pressure (MAP) - 1.4 × education level code); e = 2.718.
Collapse
Affiliation(s)
- Lu Zong
- Department of Obstetrics & Gynecology, First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | | | | | | | | |
Collapse
|
49
|
Chui A, Murthi P, Brennecke SP, Ignjatovic V, Monagle PT, Said JM. The expression of placental proteoglycans in pre-eclampsia. Gynecol Obstet Invest 2012; 73:277-84. [PMID: 22516801 DOI: 10.1159/000333262] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Accepted: 09/12/2011] [Indexed: 11/19/2022]
Abstract
BACKGROUND/AIMS Pre-eclampsia (PE) is one of the leading causes of maternal and perinatal morbidity and mortality. PE is defined clinically as the onset of maternal hypertension and proteinuria following 20 weeks of gestation. It is associated with altered maternal uterine decidual spiral artery remodelling, which may lead to reduced blood flow and increased thrombosis within the uteroplacental vasculature. Proteoglycans (PGs) are macromolecules which have (in combination with glycosaminoglycans) important anticoagulant roles in vascular endothelial environments, including the uteroplacental circulation. The hypothesis under consideration in this study was that differential expression of placental PGs may be associated with PE. METHODS PE and control placental samples were collected with ethics approval and patient consent. RNA and protein were extracted and real-time PCR and Western immunoblotting were performed to determine the expression of the PGs in the samples. RESULTS Of the nine PGs investigated, none showed increased expression, whereas the mRNA and protein expression of five of them was significantly decreased in the placentae of pre-eclamptic women compared to gestation-matched controls. CONCLUSION Therefore, the results of this study support the hypothesis that a placental PG deficiency may contribute to the placental thrombotic lesions characteristic of PE.
Collapse
Affiliation(s)
- A Chui
- Department of Perinatal Medicine, Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Vic., Australia
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
The invasion of extravillous cytotrophoblasts (EVT) into the underlying maternal tissues and vasculature is a key step in human placentation. The molecular mechanisms involved in the development of the invasive phenotype of EVT include many that were first discovered for their role in cancer cell metastasis. Previous studies have demonstrated that N-cadherin and its regulatory transcription factor Twist play important roles in the onset and progression of cancers, but their roles in human trophoblastic cell invasion is not clear. The goal of the study was to examine the role of Twist and N-cadherin in human trophoblastic cell invasion. Twist and N-cadherin mRNA and protein levels were determined by RT-PCR and Western blotting in human placental tissues, highly invasive EVT, and poorly invasive JEG-3 and BeWo cells. Whether IL-1β and TGF-β1 regulate Twist mRNA and protein levels in the EVT was also examined. A small interfering RNA strategy was employed to determine the role of Twist and N-cadherin in HTR-8/SVneo cell invasion. Matrigel assays were used to assess cell invasion. Twist and N-cadherin were highly expressed in EVT but were poorly expressed in JEG-3 and BeWo cells. IL-1β and TGF-β1 differentially regulated Twist expression in EVT in a time- and concentration-dependent manner. Small interfering RNA specific for Twist decreased N-cadherin and reduced invasion of HTR-8/SVneo cells. Similarly, a reduction in N-cadherin decreased the invasive capacity of HTR-8/SVneo cells. Twist is an upstream regulator of N-cadherin-mediated invasion of human trophoblastic cells.
Collapse
Affiliation(s)
- York Hunt Ng
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, British Columbia, Canada V6H 3V5
| | | | | |
Collapse
|